EP4083042A1 - Spiro ring-containing quinazoline compound - Google Patents

Spiro ring-containing quinazoline compound Download PDF

Info

Publication number
EP4083042A1
EP4083042A1 EP20905322.2A EP20905322A EP4083042A1 EP 4083042 A1 EP4083042 A1 EP 4083042A1 EP 20905322 A EP20905322 A EP 20905322A EP 4083042 A1 EP4083042 A1 EP 4083042A1
Authority
EP
European Patent Office
Prior art keywords
alkyl
compound
alkoxy
cycloalkyl
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP20905322.2A
Other languages
German (de)
French (fr)
Inventor
Yuli Xie
Houxing Fan
Gang Cao
Lihui QIAN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wigen Biomedicine Technology Shanghai Co Ltd
Original Assignee
Wigen Biomedicine Technology Shanghai Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from CN201911386239.6A external-priority patent/CN113045570A/en
Priority claimed from CN202010486384.8A external-priority patent/CN113754659A/en
Application filed by Wigen Biomedicine Technology Shanghai Co Ltd filed Critical Wigen Biomedicine Technology Shanghai Co Ltd
Publication of EP4083042A1 publication Critical patent/EP4083042A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present invention belongs to the field of medicinal chemistry, and particularly to a spiro ring-containing quinazoline compound, a preparation method therefor, and use of the compound as a K-Ras G12C inhibitor in preparing antitumor medicaments.
  • Ras protein family members are important signal transduction molecules in cells, and play an important role in the growth and development. Extensive analysis and study of in vitro tumor cells, animal models and human tumor samples indicate that the over-activation of Ras family proteins is an early event in the development of human tumors and is one of the important causes of the development and progression of many types of cancer. Targeting Ras proteins and inhibiting the Ras protein activity are therefore important means of treating related tumors.
  • Ras protein exists in two forms. It is in an unactivated resting state when bound to GDP, and when a cell receives signals such as growth factor stimulation, it is bound to GTP and thus activated. Activated Ras proteins recruit a variety of signal-transducing adaptor proteins to promote phosphorylation of downstream signaling molecules such as ERK and S6, thereby activating the Ras signal transduction pathway and regulating the growth, survival, migration and differentiation of cells. Ras proteins can hydrolyze GTP back to GDP due to their GTPase activity. Besides, the GTPase-activating proteins (GAPs) in cells interact with Ras, greatly improving the GTPase activity of Ras and thereby preventing Ras proteins from being overly activated.
  • GAPs GTPase-activating proteins
  • Mutations in the K-Ras, H-Ras and N-Ras proteins of the Ras protein family are one of the common genetic mutations in a variety of tumors, and are a major factor leading to over-activation of Ras proteins in tumors.
  • Ras proteins with these mutations have unregulated activity; they are stably bound to GTP and constantly activated, thereby promoting the growth, migration and differentiation of tumor cells.
  • those in K-Ras proteins are the most common ones, accounting for 85% of all Ras mutations, while those in N-Ras (12%) and H-Ras (3%) are relatively rare.
  • K-Ras mutations are very common in many types of cancer, including pancreatic cancer (95%), colorectal cancer (45%), lung cancer (25%), etc., while relatively rare ( ⁇ 2%) in breast cancer, ovarian cancer and brain cancer.
  • K-Ras mutations mainly occur at position G12, and G12C mutation is the most common one.
  • NSCLC non-small cell lung cancer
  • K-Ras G12C K-Ras G12C
  • G12V and G12D are the second most common mutations.
  • K-Ras mutations in non-small cell lung cancer generally do not coexist with EGFR, ALK, ROS1, RET and BRAF mutations, but coexist with STK11, KEAP1, TP53 and other mutations, suggesting that K-Ras mutations may be involved in malignant transformation, proliferation and invasion of cells synergistically with STK11, KEAP1, TP53 and other mutations.
  • abnormal activation of Ras proteins is also involved in non-tumor diseases including diabetes, neurodegenerative diseases, etc.
  • Ras protein-targeting small-molecule compounds can benefit a large number of cancer patients with specific genetic mutations and non-cancer patients with over-activation of the Ras pathway.
  • K-Ras G12C muteins as a leading therapeutic target, have not been extensively researched at present, and only a few compounds, such as AMG510 of Amgen and MRTX849 of Mirati, have been under clinical research.
  • a K-Ras G12C mutation-targeting covalent inhibitor ARS-1620 was reported in Cell ( Cell, 2018, 172: 578-589 ).
  • the present invention aims to provide a compound of a structural general formula as shown in formula (1), isomers thereof, crystalline forms thereof, pharmaceutically acceptable salts thereof, hydrates thereof or solvates thereof: wherein in formula (1):
  • R 1 is H, F, Cl, Me, Et, vinyl, isopropyl, ethynyl or cyclopropyl.
  • R 2 is CH 3 CH 2 O-, CF 3 CH 2 O-, CHF 2 CH 2 O-,
  • R 3 is
  • R 4 is H, F, CN, Me, CF 3 ,
  • R 5 is:
  • R 3 when R 3 is and R 4 is F, CN, Me, CF 3 , or when R 3 is
  • R 5 is:
  • a representative compound of general formula (1) of the present invention has one of the following structures:
  • Another aspect of the present invention aims to provide a compound with a structural general formula as shown in formula (2), isomers thereof, crystalline forms thereof, pharmaceutically acceptable salts thereof, hydrates thereof or solvates thereof: wherein in formula (2):
  • R 3a is
  • R 5a is: H, or
  • a representative compound of general formula (2) of the present invention has one of the following structures:
  • Another aspect of the present invention aims to provide a compound with a structure as shown in general formula (3), isomers thereof, crystalline forms thereof, pharmaceutically acceptable salts thereof, hydrates thereof or solvates thereof:
  • R 5b is: H
  • Another purpose of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable excipient or carrier, and the compounds of general formulas (1) through (3), the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof of the present invention as active ingredients.
  • Still another purpose of the present invention is to provide use of the compounds, the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof of the present invention described above in preparing a medicament for treating RAS-associated diseases.
  • the inventors found that in the compounds of general formulas (1) through (3), when R 5 (or R 5a or R 5b ) is a spiro ring or other substituted heterocyclic ring, the compounds have very high K-RAS G12C inhibitory activity, meanwhile, the pharmacokinetic properties of the compounds are greatly improved, and the in vivo activity of the compounds is enhanced.
  • the inventors found that when position 2 (substituent R 4 ) of acrylamide is substituted with a F atom that is small in size, the compounds also have good K-RAS G12C inhibitory activity and pharmacokinetic properties.
  • the compounds of general formulas (1) through (3) described above may be synthesized using standard synthetic techniques or well-known techniques in combination with the methods described herein. In addition, solvents, temperatures and other reaction conditions mentioned herein may vary. Starting materials for the synthesis of the compounds may be obtained synthetically or commercially. The compounds described herein and other related compounds having different substituents may be synthesized using well-known techniques and starting materials, including the methods found in March, ADVANCED ORGANIC CHEMISTRY, 4th Ed., (Wiley 1992 ); Carey and Sundberg, ADVANCED ORGANIC CHEMISTRY, 4th Ed., Vols.
  • the compounds described herein are prepared according to methods well known in the art. However, the conditions involved in the methods, such as reactants, solvent, base, amount of the compound used, reaction temperature and time required for the reaction are not limited to the following explanation.
  • the compounds of the present invention can also be conveniently prepared by optionally combining various synthetic methods described herein or known in the art, and such combinations can be easily determined by those skilled in the art to which the present invention pertains.
  • the present invention also provides a method for preparing the compounds of general formulas (1) through (3), which are prepared using general reaction scheme 1 below:
  • the preparation may be performed according to general reaction scheme 1, wherein T represents H, F, Cl or I, T 1 represents R 5 , R 5a or R 5b , T 2 represents R 3 or R 3a , T 3 represents R 1 or R 1a , T 4 represents R 2 or R 2a , and T 5 represents R 4 or R 4a ; R 1 , R 1a , R 2 , R 2a , R 3 , R 3a , R 4 , R 4a , R 5 , R 5a and R 5b are defined as above, PG represents a protecting group, and X represents boric acid, a borate or a trifluoroborate.
  • “Pharmaceutically acceptable” herein refers to a substance, such as a carrier or diluent, which will not cause a compound to lose its biological activity or properties. It is relatively non-toxic; for example, when an individual is given a substance, it will not cause unwanted biological effects or interact with any component contained therein in a deleterious manner.
  • pharmaceutically acceptable salt refers to a form of a compound that does not cause significant irritation to the organism for drug administration or eliminate the biological activity and properties of the compound.
  • pharmaceutically acceptable salts are obtained by reacting the compounds of general formulas (1) through (3) with acids, e.g.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, phosphoric acid and nitric acid
  • organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, trifluoroacetic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, benzenesulfonic acid and p -toluenesulfonic acid
  • acidic amino acids such as aspartic acid and glutamic acid.
  • references to pharmaceutically acceptable salts include solvent addition forms or crystal forms, especially solvates or polymorphs.
  • a solvate contains either stoichiometric or non-stoichiometric amount of solvent and is selectively formed during crystallization with pharmaceutically acceptable solvents such as water and ethanol. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is ethanol.
  • the solvates of the compounds of general formulas (1) through (3) are conveniently prepared or formed according to the methods described herein.
  • the hydrates of the compounds of general formulas (1) through (3) are conveniently prepared by recrystallization from a mixed solvent of water/organic solvent, wherein the organic solvent used includes, but is not limited to, tetrahydrofuran, acetone, ethanol or methanol.
  • the compounds mentioned herein can exist in both non-solvated and solvated forms. In general, the solvated forms are considered equivalent to the non-solvated forms for purposes of the compounds and methods provided herein.
  • the compounds of general formulas (1) through (3) are prepared into different forms, including but not limited to amorphous, pulverized and nanoparticle forms.
  • the compound of general formula (1) includes crystalline forms, and may also be polymorphs. Polymorphs include different lattice arrangements of the same elements of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystalline forms, optical and electrical properties, stability and solubility. Different factors such as recrystallization solvent, crystallization rate and storage temperature may lead to monocrystalline form being dominant.
  • the compounds of general formulas (1) through (3) have axial chiralities and/or chiral centers and thus occur in the form of a racemate, racemic mixture, single enantiomer, diastereomeric compound and single diastereomer.
  • Each of these axial chiralities will independently produce two optical isomers, and all possible optical isomers, diastereomeric mixtures and pure or partially pure compounds are included within the scope of the present invention.
  • the present invention is meant to include all such isomeric forms of these compounds.
  • alkyl refers to a saturated aliphatic hydrocarbon group, including linear and branched groups containing 1 to 6 carbon atoms. Lower alkyl containing 1 to 4 carbon atoms, such as methyl, ethyl, propyl, 2-propyl, n -butyl, isobutyl or tert-butyl, is preferred.
  • alkyl includes unsubstituted and substituted alkyl, particularly alkyl substituted with one or more halogens. Preferred alkyl is selected from CH 3 , CH 3 CH 2 , CF 3 , CHF 2 , CF 3 CH, i Pr, n Pr, i Bu, n Bu and t Bu.
  • alkenyl refers to an unsaturated aliphatic hydrocarbon group containing carbon-carbon double bonds, including linear and branched groups containing 1 to 6 carbon atoms. Lower alkenyl containing 1 to 4 carbon atoms, such as vinyl, 1-propenyl, 1-butenyl or 2-methylpropenyl, is preferred.
  • alkynyl refers to an unsaturated aliphatic hydrocarbon group containing carbon-carbon triple bonds, including linear and branched groups containing 1 to 6 carbon atoms. Lower alkenyl containing 1 to 4 carbon atoms, such as ethynyl, 1-propynyl or 1-butynyl, is preferred.
  • cycloalkyl refers to a 3- to 6-membered all-carbon monocyclic aliphatic hydrocarbon group, wherein one or more of the rings may contain one or more double bonds, but none of them has a fully conjugated ⁇ -electron system.
  • cyclopropyl, cyclobutyl, cyclopentyl, cyclohexane, and cyclohexadiene are examples of compounds that are commonly known as cyclobutyl.
  • alkoxy refers to an alkyl group that bonds to the rest of the molecule through an ether oxygen atom.
  • Representative alkoxy groups are ones having 1-6 carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec -butoxy and tert -butoxy.
  • alkoxy includes unsubstituted and substituted alkoxy, particularly alkoxy substituted with one or more halogens.
  • Preferred alkoxy is selected from OCH 3 , OCF 3 , CHF 2 O, CF 3 CH 2 O, i- PrO, n- PrO, i- BuO, n- BuO and t- BuO.
  • heteroaryl refers to an aromatic group containing one or more heteroatoms (O, S or N) and it is monocyclic or polycyclic; for example, a monocyclic heteroaryl ring fuses with one or more carbocyclic aromatic groups or other monocyclic heterocyclyl groups.
  • heteroaryl examples include, but are not limited to, pyridyl, pyridazinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, indolyl, benzimidazolyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, benzopyridyl, and pyrrolopyrimidinyl.
  • heterocycloalkyl refers to a saturated or partially unsaturated ring system group containing one or more heteroatoms (O, S or N), wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom is optionally quaternized as a ring atom.
  • heterocycloalkyl ring system may be a monocyclic, bicyclic, spiro or polycyclic ring system.
  • Heterocycloalkyl may link to the rest of the molecule through one or more ring carbons or heteroatoms.
  • heterocycloalkyl examples include, but are not limited to, pyrrolidine, piperidine, N -methylpiperidine, tetrahydroimidazole, pyrazolidine, butyrolactam, valerolactam, imidazolidinone, hydantoin, dioxolane, phthalimide, pyrimidine-2,4(1 H ,3 H ) -dione, 1,4-dioxane, morpholine, thiomorpholine, thiomorpholine- S -oxide, thiomorpholine- S , S -oxide, piperazine, pyran, pyridone, 3-pyrroline, thiopyran, pyrone, tetrahydrofuran, tetrahydrothiophene, quinuclidine, 2-azaspiro[3.3]heptane, etc.
  • halogen refers to fluorine, chlorine, bromine, or iodine.
  • halo or halogenated
  • the term “halo” (or “halogenated”) before a group name indicates that the group is partially or fully halogenated, that is, substituted in any combination by F, Cl, Br or I, preferably by F or Cl.
  • acceptable means that a formula component or an active ingredient does not unduly adversely affect a general therapeutic target's health.
  • treatment include alleviating, inhibiting, or ameliorating a symptom or condition of a disease; inhibiting the development of complications; ameliorating or preventing underlying metabolic syndrome; inhibiting the development of the disease or symptom, e.g., controlling the progression of the disease or condition; alleviating the disease or symptom; causing the disease or symptom to subside; alleviating a complication caused by the disease or symptom, or preventing or treating a sign caused by the disease or symptom.
  • a compound or pharmaceutical composition when administered, can ameliorate a disease, symptom, or condition, particularly meaning ameliorating the severity, delaying the onset, slowing the progression, or reducing the duration of the disease. Fixed or temporary administration, or continuous or intermittent administration, may be attributed to or associated with the administration.
  • the “active ingredient” refers to compounds of general formulas (1) through (3), and pharmaceutically acceptable inorganic or organic salts of the compounds of general formulas (1) through (3).
  • the compounds of the present invention may contain one or more asymmetric centers (axial chirality) and thus occur in the form of a racemate, racemic mixture, single enantiomer, diastereomeric compound and single diastereomer.
  • Asymmetric centers that may be present depend on the nature of the various substituents on the molecule. Each of these asymmetric centers will independently produce two optical isomers, and all possible optical isomers, diastereomeric mixtures and pure or partially pure compounds are included within the scope of the present invention.
  • the present invention is meant to include all such isomeric forms of these compounds.
  • composition refers to a compound or composition that, when administered to an individual (human or animal), is capable of inducing a desired pharmacological and/or physiological response by local and/or systemic action.
  • administering refers herein to the direct administration of the compound or composition, or the administration of a prodrug, derivative, analog or the like of the active compound.
  • the present invention provides a method for using the compound or pharmaceutical composition of the present invention to treat diseases, including but not limited to conditions involving G12C K-Ras, G12C H-Ras and/or G12C N-Ras mutations (e.g., cancer).
  • diseases including but not limited to conditions involving G12C K-Ras, G12C H-Ras and/or G12C N-Ras mutations (e.g., cancer).
  • a method for treating cancer comprising administering to an individual in need thereof an effective amount of a pharmaceutical composition of any of the aforementioned compounds of structural general formulas (1) through (3) protected.
  • the cancer is mediated by K-Ras, H-Ras and/or G12C N-Ras mutations.
  • the cancer is lung cancer, pancreatic cancer, colon cancer, MYH-associated polyposis, or colorectal cancer.
  • the compound and the pharmaceutically acceptable salt thereof of the present invention can be prepared into various preparations which include the compound or the pharmaceutically acceptable salt thereof disclosed herein in a safe and effective amount range and a pharmaceutically acceptable excipient or carrier, wherein the "safe and effective amount” means that the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the safe and effective amount of the compound is determined according to the age, condition, course of treatment and other specific conditions of a treated subject.
  • pharmaceutically acceptable excipient or carrier refers to one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity.
  • Cosmetic means that the components of the composition are capable of intermixing with the compound of the present invention and with each other, without significantly diminishing the pharmaceutical efficacy of the compound.
  • Examples of pharmaceutically acceptable excipients or carriers are cellulose and its derivatives (e.g., sodium carboxymethylcellulose, sodium ethylcellulose or cellulose acetate), gelatin, talc, solid lubricants (e.g., stearic acid or magnesium stearate), calcium sulfate, vegetable oil (e.g., soybean oil, sesame oil, peanut oil or olive oil), polyols (e.g., propylene glycol, glycerol, mannitol or sorbitol), emulsifiers (e.g., Tween ® ), wetting agents (e.g., sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives e.g., sodium carboxymethylcellulose, sodium ethylcellulose or cellulose acetate
  • gelatin talc
  • solid lubricants e.g
  • the compound of the present invention When the compound of the present invention is administered, it may be administered orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously) or topically.
  • Solid dosage forms for oral administration include capsules, tablets, pills, pulvises and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, such as starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, such as hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, such as glycerol; (d) disintegrants, such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate; (e) solution retarders, such as paraffin; (f) absorption accelerators, such as quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol, such
  • Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may include opacifying agents, and the active compound or compound in such a composition may be released in a certain part of the digestive tract in a delayed manner. Examples of embedding components that can be used are polymeric substances and wax-based substances. If necessary, the active compound can also be in microcapsule form with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage form may include inert diluents commonly used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butanediol, dimethylformamide, and oils, especially cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, or mixtures of these substances.
  • inert diluents commonly used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butanediol, dimethylformamide
  • the composition may also include adjuvants, such as wetting agents, emulsifiers, suspending agents, sweeteners, flavoring agents, and perfuming agents.
  • adjuvants such as wetting agents, emulsifiers, suspending agents, sweeteners, flavoring agents, and perfuming agents.
  • Suspensions in addition to the active compound, may include suspending agents, such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methylate and agar, or mixtures of these substances.
  • compositions for parenteral injection may include physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for redissolving into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • Dosage forms for topical administration of the compound of the present invention include ointments, pulvises, patches, sprays and inhalants.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers or propellants that may be required if necessary.
  • the compound of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
  • a safe and effective amount of the compound of the present invention is administered to a mammal (such as a human) to be treated, wherein the administration dose is a pharmaceutically effective administration dose.
  • a mammal such as a human
  • the daily dose of administration is usually 1-2000 mg, preferably 50-1000 mg.
  • factors as the route of administration, the health condition of the patient and the like will also be considered, which are well known to skilled physicians.
  • the present invention uses the following abbreviations: CD 3 OD for deuterated methanol; MeCN for acetonitrile; DCM for dichloromethane; DIPEA for diisopropylethylamine; dioxane for 1,4-dioxane; DMF for dimethylformamide; K 3 PO 4 for potassium phosphate; min for minute; MS for mass spectroscopy; NaH for sodium hydride; NMR for nuclear magnetic resonance; Pd 2 (dba) 3 for tris(dibenzylideneacetone)dipalladium; Pd(dppf)Cl 2 for [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride; TFA (CF 3 COOH) for trifluoroacetic acid; TLC for thin layer chromatography; THF for tetrahydrofuran; and Xantphos for 4,5-bis(diphenylphosphane)-9,9-dimethylxanthen
  • FIG. 1 shows the inhibition of the phosphorylated ERK (pERK) level in cells by compounds.
  • Example 1 Synthesis of 1-(7-(6-Cyclopropyl-8-ethoxy-2-((1-(2-methoxyethyl)piperidin-4-yl)oxy)-7-(5-methyl-1 H -indazol-4-yl)quinazolin-4-yl)-2,7-diazaspiro[3.5]nonan-2-yl)-2-fluoroprop-2-en-1-one (Compound 1)
  • Trifluoroethanol (0.9 g, 8.4 mmol) was dissolved in anhydrous DMF (10 mL). NaH was added under ice bath. The mixture was stirred at room temperature for 5 min to obtain sodium trifluoroethoxide.
  • Compound 1-4 (4.1 g, 5.6 mmol) was dissolved in anhydrous THF (40 mL). The solution of sodium trifluoroethoxide in DMF prepared above was added. The mixture was stirred at room temperature overnight. After the reaction was completed, water was added, followed by EA for extraction. The organic phase was dried and concentrated, and the residue was subjected to column chromatography to obtain compound 1-5 (4.5 g, 99% yield).
  • the target compound 2-341 was obtained using different starting materials according to a synthesis method similar to that in Example 1.
  • Table 1 Compound Compound structure [M+H] + Compound Compound structure [M+H] + 2 698.4 3 734.4 4 684.4 5 738.3 6 792.4 7 747.4 8 691.4 9 727.4 10 694.3 11 712.3 12 728.3 13 708.3 14 722.4 15 736.4 16 718.3 17 748.4 18 759.3 19 802.3 20 812.4 21 812.4 22 800.4 23 801.4 24 799.4 25 801.4 26 706.3 27 734.4 28 767.4 29 786.3 30 770.4 31 766.4 32 752.3 33 756.3 34 756.3 35 734.4 36 756.3 37 738.3 38 756.3 39 735.4 40 753.3 41 761.3 42 779.3 43 762.3 44 780.3 45 714.3 46 732.3 47 713.4 48 731.3 49 781.3 50 799.2 51 698.3 52 71
  • the compounds of the present application may have axial chirality. Compounds with axial chirality can be resolved to obtain two chiral isomers.
  • a first chiral isomer 174-a; retention time on the chromatography column: 8.994 min; and a second chiral isomer: 174-b; retention time on the chromatography column: 14.583 min.
  • 270 Same as those for compound 142 except that the mobile phase is ethanol- n -hexane (30/70) a first chiral isomer: 270-a; retention time on the chromatography column: 7.280 min; and a second chiral isomer: 270-b; retention time on the chromatography column: 12.962 min.
  • H358 cells were seeded in a 24-well plate. After one day of growth, a test compound (at a concentration of 1 ⁇ M) was added. After 24 h of action of the compound, the cells were lysed, and the cell lysate was transferred to a 96-well ELISA plate. The levels of pERK and ERK in the lysate were measured using an ELISA kit (abcam 176660). The ratio of pERK to ERK was calculated and compared with that of the DMSO group, and the percentage of inhibition of pERK activity by the compound was calculated. The results are shown in Table 3 below. Table 3.
  • Antiproliferative activity of the compounds of the present invention against H358 cells Compound IC 50 Compound IC 50 Compound IC 50 1 +++ 2 +++ 3 +++ 4 +++ 5 +++ 6 +++ 7 ++ 8 ++ 9 +++ 10 ++ 11 +++ 12 +++ 13 ++ 14 ++ 15 +++ 16 ++ 17 +++ 18 ++ 19 ++ 20 +++ 21 +++ 22 ++ 23 +++ 24 ++ 25 +++ 26 ++ 27 ++ 28 ++ 29 +++ 30 +++ 31 +++ 32 +++ 33 +++ 34 +++ 35 +++ 36 +++ 37 ++ 38 ++ 39 +++ 40 +++ 41 +++ 42 ++ 43 +++ 44 +++ 45 +++ 46 +++ 47 +++ 48 ++ 49 +++ 50 +++ 51 +++ 52 +++ 53 +++ 54 +++ 55 +++ 56 +++ 57 +++ 58 +++ 59 +++ 60 +++ 61 +++ 62 +++ 63 +++ 64 ++ 65 +++ 66 67 ++ 68 ++ 69 +++ 70 ++ 71
  • the antiproliferative activity of most of the compounds of the present invention against H358 cells is less than 0.3 ⁇ M, and when R 5 (or R 5a or R 5b ) is a spiro ring or other substituted heterocyclic ring, the compounds have very high K-RAS G12C inhibitory activity.
  • Compounds 131, 142 and 171 all have good antiproliferative activity against H358 cells, with their IC 50 values being 1.5 nM, 2.5 nM and 1.4 nM, respectively, while the IC 50 values of the reference compounds B and C were 4.6 nM and 5.1 nM, respectively, indicating that the cell activity of the compounds was greatly improved after cyclization of the amino groups on the side chains of the compounds.
  • the compounds when position 2 (substituent R 4 ) of acrylamide is substituted with a F atom that is small in size, the compounds also have very high K-RAS G12C inhibitory activity.
  • the compounds were administered by intravenous injection at a dose of 2 mg/kg and oral gavage at a dose of 10 mg/kg (0.5% CMC-Na suspension).
  • 15 male ICR mice were selected for each group, and each mouse was subjected to blood collection at 3 discrete time points, with 3 mice per time point.
  • the time points of sampling were as follows: before the administration, and at 5 min, 15 min, 30 min, 1 h, 3 h, 5 h, 8 h, 12 h and 24 h after the administration. 80 ⁇ L of blood was collected from the eye sockets or the hearts of the mice at each of the time points after the administration.
  • compound 131 has good oral absorption properties, and has improved metabolic parameters such as half-life (t 1 ⁇ 2 ), maximum plasma concentration (C max ), area under the drug-time curve (AUC 0-t ), and oral bioavailability.
  • compound 171 has better metabolic parameters
  • compound 142 also has significantly improved metabolic parameters such as C max and AUC 0-t , indicating that the metabolic properties of the compound are well improved after the amino groups on the side chain are cyclized.
  • the metabolic properties of the compounds similar to compounds 131 and 171 in the present application are also significantly improved. Good oral absorption properties are of great significance in improving the efficacy of drugs, reducing the dose of administration and reducing the costs.
  • Mia PaCa-2 cells were cultured conventionally in 1640 medium containing 10% fetal bovine serum in a 37 °C/5% CO 2 incubator. After passage, the cells were collected when they reached the desired amount. 1 ⁇ 10 7 Mia PaCa-2 cells were injected into the left dorsal side of each nude mouse, and the animals were randomly grouped for administration after tumors grew to 150 mm3.
  • the groups are as follows: 1) a solvent control group of 8 mice; and 2) compound 1 group, compound 2 group, compound 5 group, compound 31 group, compound 131 group, compound 142 group, compound 171 group, compound B group and compound C group, with 8 mice per group.
  • mice in the solvent control group were subjected to intragastric administration of 0.5% CMC-Na once daily; mice in compound 1 group, compound 2 group, compound 5 group, compound 31 group, compound 131 group, compound 142 group, compound 171 group, compound B group and compound C group were subjected to intragastric administration of a suspension of a compound in 0.5% CMC-Na once daily.
  • mice in compound 1 group, compound 2 group, compound 5 group, compound 31 group, compound 131 group, compound 142 group, compound 171 group, compound B group and compound C group were subjected to intragastric administration of a suspension of a compound in 0.5% CMC-Na once daily.
  • tumor volumes and body weight of the mice were measured, and the nude mice were sacrificed on day 21 of administration.
  • Table 6 Table 6.
  • the compounds of the present invention have high in vivo antitumor activity; a tumor can regress after 21 consecutive days of administration at 10 mg/kg/day; compounds 1, 5, 31, 131, 142 and 171 have higher in vivo activity than reference compound B and compound C, and compounds 142 and 171 have significantly higher in vivo activity than compound C, indicating the in vivo activity of the compound is also greatly improved after the amino groups on the side chain of the compound are cyclized.
  • H358 cells were plated on to a 24-well plate at 2 ⁇ 10 5 cells/well. Serially diluted compounds including AMG510, MRTX849, compound 142 and compound 171 were added. After overnight incubation, cells were lysed, and proteins were quantified and subjected to gel electrophoresis.
  • FIG. 1 The results of the phosphorylated ERK (pERK) level assay by western blot are shown in FIG. 1 .
  • the compounds 142 and 171 of the present invention shows stronger inhibition of the phosphorylated ERK (pERK) level in cells than the reference drugs AMG510 and MRTX849 when at the same concentration.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present invention relates to a spiro ring-containing quinazoline compound, a preparation method therefor, and use of the compound as a K-Ras G12C inhibitor in preparing antitumor medicaments.

Description

  • The present application claims priority to Chinese Patent application No. CN201911386239.6 filed on Dec. 27, 2019 and Chinese Patent application No. CN202010486384.8 filed on Jun. 1, 2020 , the contents of which are incorporated herein by reference in their entirety.
  • TECHNICAL FIELD
  • The present invention belongs to the field of medicinal chemistry, and particularly to a spiro ring-containing quinazoline compound, a preparation method therefor, and use of the compound as a K-Ras G12C inhibitor in preparing antitumor medicaments.
  • BACKGROUND
  • Ras protein family members are important signal transduction molecules in cells, and play an important role in the growth and development. Extensive analysis and study of in vitro tumor cells, animal models and human tumor samples indicate that the over-activation of Ras family proteins is an early event in the development of human tumors and is one of the important causes of the development and progression of many types of cancer. Targeting Ras proteins and inhibiting the Ras protein activity are therefore important means of treating related tumors.
  • An Ras protein exists in two forms. It is in an unactivated resting state when bound to GDP, and when a cell receives signals such as growth factor stimulation, it is bound to GTP and thus activated. Activated Ras proteins recruit a variety of signal-transducing adaptor proteins to promote phosphorylation of downstream signaling molecules such as ERK and S6, thereby activating the Ras signal transduction pathway and regulating the growth, survival, migration and differentiation of cells. Ras proteins can hydrolyze GTP back to GDP due to their GTPase activity. Besides, the GTPase-activating proteins (GAPs) in cells interact with Ras, greatly improving the GTPase activity of Ras and thereby preventing Ras proteins from being overly activated.
  • Mutations in the K-Ras, H-Ras and N-Ras proteins of the Ras protein family are one of the common genetic mutations in a variety of tumors, and are a major factor leading to over-activation of Ras proteins in tumors. Compared to the wild-type Ras proteins, Ras proteins with these mutations have unregulated activity; they are stably bound to GTP and constantly activated, thereby promoting the growth, migration and differentiation of tumor cells. Among these mutations, those in K-Ras proteins are the most common ones, accounting for 85% of all Ras mutations, while those in N-Ras (12%) and H-Ras (3%) are relatively rare. K-Ras mutations are very common in many types of cancer, including pancreatic cancer (95%), colorectal cancer (45%), lung cancer (25%), etc., while relatively rare (< 2%) in breast cancer, ovarian cancer and brain cancer. K-Ras mutations mainly occur at position G12, and G12C mutation is the most common one. For example, in non-small cell lung cancer (NSCLC), about 50% of K-Ras mutations are K-Ras G12C, and G12V and G12D are the second most common mutations. Genomic studies show that K-Ras mutations in non-small cell lung cancer generally do not coexist with EGFR, ALK, ROS1, RET and BRAF mutations, but coexist with STK11, KEAP1, TP53 and other mutations, suggesting that K-Ras mutations may be involved in malignant transformation, proliferation and invasion of cells synergistically with STK11, KEAP1, TP53 and other mutations. In addition to tumors, abnormal activation of Ras proteins is also involved in non-tumor diseases including diabetes, neurodegenerative diseases, etc. Hence, Ras protein-targeting small-molecule compounds can benefit a large number of cancer patients with specific genetic mutations and non-cancer patients with over-activation of the Ras pathway.
  • Since the discovery of Ras mutations in tumors that happened forty years ago, although we have gained deeper insight into the pathogenesis involving the Ras pathway, no clinically effective therapeutic approach targeting Ras proteins has yet come onto the market for a large number of patients with Ras protein mutations and over-activation of the Ras pathway. Therefore, the development of a high-activity small-molecule inhibitor targeted at Ras proteins, particularly the K-Ras G12C protein with high frequency of mutation, is of great clinical significance.
  • K-Ras G12C muteins, as a leading therapeutic target, have not been extensively researched at present, and only a few compounds, such as AMG510 of Amgen and MRTX849 of Mirati, have been under clinical research. In 2018, a K-Ras G12C mutation-targeting covalent inhibitor ARS-1620 was reported in Cell (Cell, 2018, 172: 578-589). A class of spiro compounds with K-Ras G12C activity and anti-tumor activity in mice are reported in patent WO2018/143315 , and a general formula A, a representative compound B (Example 35 in the patent) and a representative compound C (Example 65 in the patent) thereof are shown as the structures below (refer to the patent for the definitions of the symbols in the formula):
    Figure imgb0001
    Figure imgb0002
  • Currently, there is an urgent need to study and discover compounds with good K-Ras G12C activity and superior pharmacokinetic properties.
  • SUMMARY
  • The present invention aims to provide a compound of a structural general formula as shown in formula (1), isomers thereof, crystalline forms thereof, pharmaceutically acceptable salts thereof, hydrates thereof or solvates thereof:
    Figure imgb0003
    wherein in formula (1):
    • R1 is H, halogen, C1-C3 alkyl, C2-C4 alkenyl, C2-C4 alkynyl or C3-C6 cycloalkyl;
    • R2 is C1-C3 alkoxy, C1-C3 haloalkoxy or -NRaRb, wherein Ra and Rb are independently H, C1-C3 alkyl or C1-C3 haloalkyl, or Ra and Rb, together with a N atom, form a 4-7 membered heterocycloalkyl group, wherein the heterocycloalkyl group may be substituted with 1-3 halogen atoms;
    • R3is
      Figure imgb0004
      Figure imgb0005
      or
      Figure imgb0006
      , wherein Rc is H or F; Rd is H, F, Cl or Me; Re is H, F, Cl or Me; Rf is F, NH2, Me or cyclopropyl; Rx1, Rx2, Rx3, Rx4, Rx5, Rx6 and Rx7 are independently H, F, Cl, OH, OMe, NH2, CF3, C1-C3 alkyl or C3-C6 cycloalkyl;
    • R4 is H, halogen, CN, C1-C3 alkyl, C1-C3 haloalkyl or heteroaryl; and
    • when R3 is
      Figure imgb0007
      or
      Figure imgb0008
      , and R4 is H, R5is:
      Figure imgb0009
      Figure imgb0010
      Figure imgb0011
      Figure imgb0012
      or
      Figure imgb0013
      wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rh is
      Figure imgb0014
      Figure imgb0015
      Figure imgb0016
      Figure imgb0017
    • when R3 is
      Figure imgb0018
      and R4 is halogen, CN, C1-C3 alkyl, C1-C3 haloalkyl or heteroaryl; or, when R3 is
      Figure imgb0019
      Figure imgb0020
      is:
      Figure imgb0021
      Figure imgb0022
      Figure imgb0023
      Figure imgb0024
      or
      Figure imgb0025
      wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2;
    • Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rh is
      Figure imgb0026
      Figure imgb0027
      Figure imgb0028
      ; Ri is H, halogen, methyl or cyano.
  • In another preferred embodiment, in the general formula (1), R1 is H, F, Cl, Me, Et, vinyl, isopropyl, ethynyl or cyclopropyl.
  • In another preferred embodiment, in the general formula (1), R2 is CH3CH2O-, CF3CH2O-, CHF2CH2O-,
    Figure imgb0029
  • In another preferred embodiment, in the general formula (1), R3 is
    Figure imgb0030
    Figure imgb0031
    Figure imgb0032
    Figure imgb0033
    Figure imgb0034
    Figure imgb0035
  • In another preferred embodiment, in the general formula (1), R4 is H, F, CN, Me, CF3,
    Figure imgb0036
    Figure imgb0037
  • In another preferred embodiment, in the general formula (1), when R3 is
    Figure imgb0038
    Figure imgb0039
    and R4 is H, R5 is:
    Figure imgb0040
    Figure imgb0041
    Figure imgb0042
    Figure imgb0043
    Figure imgb0044
    Figure imgb0045
    Figure imgb0046
    Figure imgb0047
    Figure imgb0048
    Figure imgb0049
    Figure imgb0050
    Figure imgb0051
    Figure imgb0052
    Figure imgb0053
    Figure imgb0054
    Figure imgb0055
    Figure imgb0056
    Figure imgb0057
    Figure imgb0058
    Figure imgb0059
    Figure imgb0060
    Figure imgb0061
    Figure imgb0062
    Figure imgb0063
    Figure imgb0064
    Figure imgb0065
    Figure imgb0066
  • In another preferred embodiment, in the general formula (1), when R3 is
    Figure imgb0067
    Figure imgb0068
    and R4 is F, CN, Me, CF3,
    Figure imgb0069
    Figure imgb0070
    or when R3 is
    Figure imgb0071
    Figure imgb0072
    Figure imgb0073
    Figure imgb0074
    Figure imgb0075
    Figure imgb0076
    Figure imgb0077
    Figure imgb0078
    Figure imgb0079
    Figure imgb0080
    Figure imgb0081
    Figure imgb0082
    Figure imgb0083
    Figure imgb0084
    Figure imgb0085
    Figure imgb0086
    Figure imgb0087
    Figure imgb0088
    Figure imgb0089
    Figure imgb0090
    Figure imgb0091
    Figure imgb0092
    Figure imgb0093
    Figure imgb0094
    Figure imgb0095
    Figure imgb0096
    Figure imgb0097
    Figure imgb0098
    Figure imgb0099
    Figure imgb0100
    Figure imgb0101
    Figure imgb0102
    Figure imgb0103
    Figure imgb0104
  • In another preferred embodiment, in the general formula (1), R5 is:
    Figure imgb0105
    Figure imgb0106
    Figure imgb0107
    Figure imgb0108
    Figure imgb0109
    Figure imgb0110
    Figure imgb0111
    Figure imgb0112
    Figure imgb0113
    Figure imgb0114
    Figure imgb0115
    Figure imgb0116
    Figure imgb0117
    Figure imgb0118
    Figure imgb0119
  • In various embodiments, a representative compound of general formula (1) of the present invention has one of the following structures:
    Figure imgb0120
    Figure imgb0121
    Figure imgb0122
    Figure imgb0123
    Figure imgb0124
    Figure imgb0125
    Figure imgb0126
    Figure imgb0127
    Figure imgb0128
    Figure imgb0129
    Figure imgb0130
    Figure imgb0131
    Figure imgb0132
    Figure imgb0133
    Figure imgb0134
    Figure imgb0135
    Figure imgb0136
    Figure imgb0137
    Figure imgb0138
    Figure imgb0139
    Figure imgb0140
    Figure imgb0141
    Figure imgb0142
    Figure imgb0143
    Figure imgb0144
    Figure imgb0145
    Figure imgb0146
    Figure imgb0147
    Figure imgb0148
    Figure imgb0149
    Figure imgb0150
    Figure imgb0151
    Figure imgb0152
    Figure imgb0153
    Figure imgb0154
    Figure imgb0155
    Figure imgb0156
    Figure imgb0157
    Figure imgb0158
    Figure imgb0159
    Figure imgb0160
    Figure imgb0161
    Figure imgb0162
    Figure imgb0163
    Figure imgb0164
    Figure imgb0165
    Figure imgb0166
    Figure imgb0167
    Figure imgb0168
    Figure imgb0169
    Figure imgb0170
    Figure imgb0171
    Figure imgb0172
    Figure imgb0173
    Figure imgb0174
    Figure imgb0175
    Figure imgb0176
    Figure imgb0177
    Figure imgb0178
    Figure imgb0179
    Figure imgb0180
    Figure imgb0181
    Figure imgb0182
    Figure imgb0183
    Figure imgb0184
    Figure imgb0185
    Figure imgb0186
    Figure imgb0187
    Figure imgb0188
    Figure imgb0189
    Figure imgb0190
    Figure imgb0191
    Figure imgb0192
    Figure imgb0193
    Figure imgb0194
  • Another aspect of the present invention aims to provide a compound with a structural general formula as shown in formula (2), isomers thereof, crystalline forms thereof, pharmaceutically acceptable salts thereof, hydrates thereof or solvates thereof:
    Figure imgb0195
    wherein in formula (2):
    • R1a is
      Figure imgb0196
    • R2a is CH3O-, CH3CH2O-, CF3CH2O- or CHF2CH2O-;
    • R3a is
      Figure imgb0197
      Figure imgb0198
      wherein Rc is H or F, Rd is H, F, Cl or Me, Re is H, F, Cl or Me, and Rf is F, NH2, Me or cyclopropyl;
    • R4a is H or F; and
    • R5a is: H,
      Figure imgb0199
      Figure imgb0200
      Figure imgb0201
      or
      Figure imgb0202
      , wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; v is an integer of 1, 2 or 3; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rj is independently halogen, CN, SO2Me, C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl or
      Figure imgb0203
      Rk is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy, C3-C6 cycloalkyl or
      Figure imgb0204
      Rn is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy or C3-C6 cycloalkyl, two Rn groups, together with one carbon atom, form a spiro ring, or two Rn groups, together with different carbon atoms, form a bridged ring; R1 and Rm are independently C1-C3 alkyl, C1-C3 haloalkyl, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, or R1 and Rm, together with a N atom, form a 3-8 membered heterocycloalkyl group, wherein the 3-8 membered heterocycloalkyl group may be substituted with 1-3 groups selected from OH, halogen, cyano, C1-C3 alkyl, C3-C6 cycloalkyl, heterocycloalkyl, (C1-C3)alkoxy or (halogenated C1-C3)alkoxy.
  • In another preferred embodiment, in the general formula (2), R3a is
    Figure imgb0205
    Figure imgb0206
    Figure imgb0207
  • In another preferred embodiment, in the general formula (2), R5a is: H,
    Figure imgb0208
    Figure imgb0209
    Figure imgb0210
    Figure imgb0211
    Figure imgb0212
    Figure imgb0213
    Figure imgb0214
    Figure imgb0215
    Figure imgb0216
    Figure imgb0217
    Figure imgb0218
    Figure imgb0219
    Figure imgb0220
    Figure imgb0221
    Figure imgb0222
    Figure imgb0223
    Figure imgb0224
    Figure imgb0225
    Figure imgb0226
    Figure imgb0227
    Figure imgb0228
    Figure imgb0229
    Figure imgb0230
    Figure imgb0231
    Figure imgb0232
    Figure imgb0233
    Figure imgb0234
    Figure imgb0235
    Figure imgb0236
    Figure imgb0237
    Figure imgb0238
    or
    Figure imgb0239
  • In various embodiments, a representative compound of general formula (2) of the present invention has one of the following structures:
    Figure imgb0240
    Figure imgb0241
    Figure imgb0242
    Figure imgb0243
    Figure imgb0244
    Figure imgb0245
    Figure imgb0246
    Figure imgb0247
    Figure imgb0248
    Figure imgb0249
    Figure imgb0250
    Figure imgb0251
    Figure imgb0252
    Figure imgb0253
    Figure imgb0254
    Figure imgb0255
    Figure imgb0256
    Figure imgb0257
    Figure imgb0258
    Figure imgb0259
    Figure imgb0260
    Figure imgb0261
    Figure imgb0262
    Figure imgb0263
    Figure imgb0264
    Figure imgb0265
    Figure imgb0266
    Figure imgb0267
    Figure imgb0268
    Figure imgb0269
    Figure imgb0270
    Figure imgb0271
    Figure imgb0272
    Figure imgb0273
    Figure imgb0274
    Figure imgb0275
    Figure imgb0276
    Figure imgb0277
    Figure imgb0278
  • Another aspect of the present invention aims to provide a compound with a structure as shown in general formula (3), isomers thereof, crystalline forms thereof, pharmaceutically acceptable salts thereof, hydrates thereof or solvates thereof:
    Figure imgb0279
    • wherein, R5b is:
      Figure imgb0280
      Figure imgb0281
      Figure imgb0282
      Figure imgb0283
      Figure imgb0284
      wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rh is
      Figure imgb0285
      Figure imgb0286
      Figure imgb0287
      Ri is H, halogen, methyl or cyano; or
    • R5b is: H,
      Figure imgb0288
      Figure imgb0289
      Figure imgb0290
      or
      Figure imgb0291
      wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; v is an integer of 1, 2 or 3; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rj is independently halogen, CN, SO2Me, C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl or
      Figure imgb0292
      ; Rk is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy, C3-C6 cycloalkyl or
      Figure imgb0293
      ; Rn is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy or C3-C6 cycloalkyl, two Rn groups, together with one carbon atom, form a spiro ring, or two Rn groups, together with different carbon atoms, form a bridged ring; Rl and Rm are independently C1-C3 alkyl, C1-C3 haloalkyl, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, or Rl and Rm, together with a N atom, form a 3-8 membered heterocycloalkyl group, wherein the 3-8 membered heterocycloalkyl group may be substituted with 1-3 groups selected from OH, halogen, cyano, C1-C3 alkyl, C3-C6 cycloalkyl, heterocycloalkyl, (C1-C3)alkoxy or (halogenated C1-C3)alkoxy.
  • In another preferred embodiment, in the general formula (3), R5b is: H,
    Figure imgb0294
    Figure imgb0295
    Figure imgb0296
    Figure imgb0297
    Figure imgb0298
    Figure imgb0299
    Figure imgb0300
    Figure imgb0301
    Figure imgb0302
    Figure imgb0303
    Figure imgb0304
    Figure imgb0305
    Figure imgb0306
    Figure imgb0307
    Figure imgb0308
    Figure imgb0309
    Figure imgb0310
    Figure imgb0311
    Figure imgb0312
    Figure imgb0313
    Figure imgb0314
    Figure imgb0315
    Figure imgb0316
    Figure imgb0317
    Figure imgb0318
    Figure imgb0319
    Figure imgb0320
    Figure imgb0321
    Figure imgb0322
    Figure imgb0323
    Figure imgb0324
    Figure imgb0325
    Figure imgb0326
    Figure imgb0327
    Figure imgb0328
    Figure imgb0329
    Figure imgb0330
    Figure imgb0331
    Figure imgb0332
    Figure imgb0333
    Figure imgb0334
    Figure imgb0335
    Figure imgb0336
    Figure imgb0337
    Figure imgb0338
    Figure imgb0339
    Figure imgb0340
    Figure imgb0341
    Figure imgb0342
    Figure imgb0343
    Figure imgb0344
    Figure imgb0345
    Figure imgb0346
    Figure imgb0347
    Figure imgb0348
    Figure imgb0349
    Figure imgb0350
    Figure imgb0351
    Figure imgb0352
    Figure imgb0353
    Figure imgb0354
    Figure imgb0355
    Figure imgb0356
    Figure imgb0357
    Figure imgb0358
    Figure imgb0359
    Figure imgb0360
    Figure imgb0361
    Figure imgb0362
    Figure imgb0363
    Figure imgb0364
    Figure imgb0365
    Figure imgb0366
  • Another purpose of the present invention is to provide a pharmaceutical composition comprising a pharmaceutically acceptable excipient or carrier, and the compounds of general formulas (1) through (3), the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof of the present invention as active ingredients.
  • Still another purpose of the present invention is to provide use of the compounds, the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof of the present invention described above in preparing a medicament for treating RAS-associated diseases.
  • Through synthesis of and careful studies on various new compounds with K-RAS G12C inhibitory effects, the inventors found that in the compounds of general formulas (1) through (3), when R5 (or R5a or R5b) is a spiro ring or other substituted heterocyclic ring, the compounds have very high K-RAS G12C inhibitory activity, meanwhile, the pharmacokinetic properties of the compounds are greatly improved, and the in vivo activity of the compounds is enhanced. In another aspect, the inventors found that when position 2 (substituent R4) of acrylamide is substituted with a F atom that is small in size, the compounds also have good K-RAS G12C inhibitory activity and pharmacokinetic properties.
  • It should be understood that both the above general description and the following detailed description of the present invention are exemplary and explanatory, and are intended to provide further explanation of the present invention claimed.
  • Synthesis of the Compounds
  • Methods for preparing the compounds of general formulas (1) through (3) of the present invention are hereafter described in detail, but these specific methods do not limit the present invention in any way.
  • The compounds of general formulas (1) through (3) described above may be synthesized using standard synthetic techniques or well-known techniques in combination with the methods described herein. In addition, solvents, temperatures and other reaction conditions mentioned herein may vary. Starting materials for the synthesis of the compounds may be obtained synthetically or commercially. The compounds described herein and other related compounds having different substituents may be synthesized using well-known techniques and starting materials, including the methods found in March, ADVANCED ORGANIC CHEMISTRY, 4th Ed., (Wiley 1992); Carey and Sundberg, ADVANCED ORGANIC CHEMISTRY, 4th Ed., Vols. A and B (Plenum 2000, 2001), and Green and Wuts, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS, 3rd Ed., (Wiley 1999). General methods for preparing a compound can be changed by using appropriate reagents and conditions for introducing different groups into the formulas provided herein.
  • In one aspect, the compounds described herein are prepared according to methods well known in the art. However, the conditions involved in the methods, such as reactants, solvent, base, amount of the compound used, reaction temperature and time required for the reaction are not limited to the following explanation. The compounds of the present invention can also be conveniently prepared by optionally combining various synthetic methods described herein or known in the art, and such combinations can be easily determined by those skilled in the art to which the present invention pertains. In one aspect, the present invention also provides a method for preparing the compounds of general formulas (1) through (3), which are prepared using general reaction scheme 1 below:
    Figure imgb0367
  • In an embodiment of the compound of general formula (1), the preparation may be performed according to general reaction scheme 1, wherein T represents H, F, Cl or I, T1 represents R5, R5a or R5b, T2 represents R3 or R3a, T3 represents R1 or R1a, T4 represents R2 or R2a, and T5 represents R4 or R4a; R1, R1a, R2, R2a, R3, R3a, R4, R4a, R5, R5a and R5b are defined as above, PG represents a protecting group, and X represents boric acid, a borate or a trifluoroborate. As shown in general reaction scheme 1, compound A1 (synthesized according to WO2018/143315 ) is reacted with compound A2 under basic conditions to give compound A3, compound A3 is reacted with T1H under basic conditions to give compound A4, compound A4 is reacted with T2H under basic conditions to give compound A5; when T = I, compound A5 and T3X are subjected to a coupling reaction to give compound A6, and compound A6 and T4X are subjected to another coupling reaction to give compound A7; when T = H, F or Cl, compound A5 and T4X are subjected to another coupling reaction to directly give compound A7; the protecting group is removed from compound A7 to give compound A8, and compound A8 is reacted with compound A9 to give the target compound A10.
  • Further Forms of Compounds
  • "Pharmaceutically acceptable" herein refers to a substance, such as a carrier or diluent, which will not cause a compound to lose its biological activity or properties. It is relatively non-toxic; for example, when an individual is given a substance, it will not cause unwanted biological effects or interact with any component contained therein in a deleterious manner.
  • The term "pharmaceutically acceptable salt" refers to a form of a compound that does not cause significant irritation to the organism for drug administration or eliminate the biological activity and properties of the compound. In certain specific aspects, pharmaceutically acceptable salts are obtained by reacting the compounds of general formulas (1) through (3) with acids, e.g. inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, phosphoric acid and nitric acid, organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, trifluoroacetic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, benzenesulfonic acid and p-toluenesulfonic acid, and acidic amino acids such as aspartic acid and glutamic acid.
  • It should be understood that references to pharmaceutically acceptable salts include solvent addition forms or crystal forms, especially solvates or polymorphs. A solvate contains either stoichiometric or non-stoichiometric amount of solvent and is selectively formed during crystallization with pharmaceutically acceptable solvents such as water and ethanol. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is ethanol. The solvates of the compounds of general formulas (1) through (3) are conveniently prepared or formed according to the methods described herein. For example, the hydrates of the compounds of general formulas (1) through (3) are conveniently prepared by recrystallization from a mixed solvent of water/organic solvent, wherein the organic solvent used includes, but is not limited to, tetrahydrofuran, acetone, ethanol or methanol. Furthermore, the compounds mentioned herein can exist in both non-solvated and solvated forms. In general, the solvated forms are considered equivalent to the non-solvated forms for purposes of the compounds and methods provided herein.
  • In other specific examples, the compounds of general formulas (1) through (3) are prepared into different forms, including but not limited to amorphous, pulverized and nanoparticle forms. In addition, the compound of general formula (1) includes crystalline forms, and may also be polymorphs. Polymorphs include different lattice arrangements of the same elements of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystalline forms, optical and electrical properties, stability and solubility. Different factors such as recrystallization solvent, crystallization rate and storage temperature may lead to monocrystalline form being dominant.
  • In another aspect, the compounds of general formulas (1) through (3) have axial chiralities and/or chiral centers and thus occur in the form of a racemate, racemic mixture, single enantiomer, diastereomeric compound and single diastereomer. Each of these axial chiralities will independently produce two optical isomers, and all possible optical isomers, diastereomeric mixtures and pure or partially pure compounds are included within the scope of the present invention. The present invention is meant to include all such isomeric forms of these compounds.
  • Terminology
  • Unless otherwise stated, the terms used in the present application, including those in the specification and claims, are defined as follows. It must be noted that in the specification and the appended claims, the singular forms "a" and "an" include plural meanings unless the context clearly indicates otherwise. Unless otherwise stated, conventional methods of mass spectrometry, nuclear magnetic resonance spectroscopy, HPLC, protein chemistry, biochemistry, recombinant DNA technology and pharmacology are used. In the present application, "or" or "and" is used to mean "and/or" unless otherwise stated.
  • Unless otherwise specified, "alkyl" refers to a saturated aliphatic hydrocarbon group, including linear and branched groups containing 1 to 6 carbon atoms. Lower alkyl containing 1 to 4 carbon atoms, such as methyl, ethyl, propyl, 2-propyl, n-butyl, isobutyl or tert-butyl, is preferred. As used herein, "alkyl" includes unsubstituted and substituted alkyl, particularly alkyl substituted with one or more halogens. Preferred alkyl is selected from CH3, CH3CH2, CF3, CHF2, CF3CH, iPr, nPr, iBu, nBu and tBu.
  • Unless otherwise specified, "alkenyl" refers to an unsaturated aliphatic hydrocarbon group containing carbon-carbon double bonds, including linear and branched groups containing 1 to 6 carbon atoms. Lower alkenyl containing 1 to 4 carbon atoms, such as vinyl, 1-propenyl, 1-butenyl or 2-methylpropenyl, is preferred.
  • Unless otherwise specified, "alkynyl" refers to an unsaturated aliphatic hydrocarbon group containing carbon-carbon triple bonds, including linear and branched groups containing 1 to 6 carbon atoms. Lower alkenyl containing 1 to 4 carbon atoms, such as ethynyl, 1-propynyl or 1-butynyl, is preferred.
  • Unless otherwise specified, "cycloalkyl" refers to a 3- to 6-membered all-carbon monocyclic aliphatic hydrocarbon group, wherein one or more of the rings may contain one or more double bonds, but none of them has a fully conjugated π-electron system. For example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexane, and cyclohexadiene.
  • Unless otherwise specified, "alkoxy" refers to an alkyl group that bonds to the rest of the molecule through an ether oxygen atom. Representative alkoxy groups are ones having 1-6 carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy and tert-butoxy. As used herein, "alkoxy" includes unsubstituted and substituted alkoxy, particularly alkoxy substituted with one or more halogens. Preferred alkoxy is selected from OCH3, OCF3, CHF2O, CF3CH2O, i-PrO, n-PrO, i-BuO, n-BuO and t-BuO.
  • Unless otherwise specified, "heteroaryl" refers to an aromatic group containing one or more heteroatoms (O, S or N) and it is monocyclic or polycyclic; for example, a monocyclic heteroaryl ring fuses with one or more carbocyclic aromatic groups or other monocyclic heterocyclyl groups. Examples of heteroaryl include, but are not limited to, pyridyl, pyridazinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, indolyl, benzimidazolyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, benzopyridyl, and pyrrolopyrimidinyl.
  • Unless otherwise specified, "heterocycloalkyl" refers to a saturated or partially unsaturated ring system group containing one or more heteroatoms (O, S or N), wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom is optionally quaternized as a ring atom. Unless otherwise stated, the "heterocycloalkyl" ring system may be a monocyclic, bicyclic, spiro or polycyclic ring system. "Heterocycloalkyl" may link to the rest of the molecule through one or more ring carbons or heteroatoms. Examples of "heterocycloalkyl" include, but are not limited to, pyrrolidine, piperidine, N-methylpiperidine, tetrahydroimidazole, pyrazolidine, butyrolactam, valerolactam, imidazolidinone, hydantoin, dioxolane, phthalimide, pyrimidine-2,4(1H,3H) -dione, 1,4-dioxane, morpholine, thiomorpholine, thiomorpholine-S-oxide, thiomorpholine-S,S-oxide, piperazine, pyran, pyridone, 3-pyrroline, thiopyran, pyrone, tetrahydrofuran, tetrahydrothiophene, quinuclidine, 2-azaspiro[3.3]heptane, etc.
  • Unless otherwise specified, "halogen" (or halo) refers to fluorine, chlorine, bromine, or iodine. The term "halo" (or "halogenated") before a group name indicates that the group is partially or fully halogenated, that is, substituted in any combination by F, Cl, Br or I, preferably by F or Cl.
  • Specific Pharmaceutical and Medical Terminology
  • The term "acceptable", as used herein, means that a formula component or an active ingredient does not unduly adversely affect a general therapeutic target's health.
  • The terms "treatment," "treatment course," or "therapy", as used herein, include alleviating, inhibiting, or ameliorating a symptom or condition of a disease; inhibiting the development of complications; ameliorating or preventing underlying metabolic syndrome; inhibiting the development of the disease or symptom, e.g., controlling the progression of the disease or condition; alleviating the disease or symptom; causing the disease or symptom to subside; alleviating a complication caused by the disease or symptom, or preventing or treating a sign caused by the disease or symptom. As used herein, a compound or pharmaceutical composition, when administered, can ameliorate a disease, symptom, or condition, particularly meaning ameliorating the severity, delaying the onset, slowing the progression, or reducing the duration of the disease. Fixed or temporary administration, or continuous or intermittent administration, may be attributed to or associated with the administration.
  • The "active ingredient" refers to compounds of general formulas (1) through (3), and pharmaceutically acceptable inorganic or organic salts of the compounds of general formulas (1) through (3). The compounds of the present invention may contain one or more asymmetric centers (axial chirality) and thus occur in the form of a racemate, racemic mixture, single enantiomer, diastereomeric compound and single diastereomer. Asymmetric centers that may be present depend on the nature of the various substituents on the molecule. Each of these asymmetric centers will independently produce two optical isomers, and all possible optical isomers, diastereomeric mixtures and pure or partially pure compounds are included within the scope of the present invention. The present invention is meant to include all such isomeric forms of these compounds.
  • The terms such as "compound", "composition", "agent" or "medicine or medicament" are used interchangeably herein and all refer to a compound or composition that, when administered to an individual (human or animal), is capable of inducing a desired pharmacological and/or physiological response by local and/or systemic action.
  • The term "administered, administering or administration" refers herein to the direct administration of the compound or composition, or the administration of a prodrug, derivative, analog or the like of the active compound.
  • Although the numerical ranges and parameters defining the broad scope of the present invention are approximations, the related numerical values set forth in the specific examples have been present herein as precisely as possible. Any numerical value, however, inherently contains a standard deviation necessarily resulting from certain methods of testing. Herein, "about" generally means that the actual value is within a particular value or range ± 10%, 5%, 1%, or 0.5%. Alternatively, the term "about" indicates that the actual value falls within the acceptable standard error of a mean, as considered by those skilled in the art. All ranges, quantities, values and percentages used herein (e.g., to describe an amount of a material, a length of time, a temperature, an operating condition, a quantitative ratio and the like) are to be understood as being modified by the word "about", except in the experimental examples or where otherwise explicitly indicated. Accordingly, unless otherwise contrarily stated, the numerical parameters set forth in the specification and the appended claims are all approximations that may vary as desired. At the very least, these numerical parameters should be construed as the significant digits indicated or the numerical value obtained using conventional rounding rules.
  • Unless otherwise defined in the specification, the scientific and technical terms used herein have the same meaning as commonly understood by those skilled in the art. Furthermore, the singular nouns used in the specification encompass their plural forms, unless contradicted by context; the plural nouns used also encompass their singular forms.
  • Therapeutic Use
  • The present invention provides a method for using the compound or pharmaceutical composition of the present invention to treat diseases, including but not limited to conditions involving G12C K-Ras, G12C H-Ras and/or G12C N-Ras mutations (e.g., cancer).
  • In some embodiments, a method for treating cancer is provided, the method comprising administering to an individual in need thereof an effective amount of a pharmaceutical composition of any of the aforementioned compounds of structural general formulas (1) through (3) protected. In some embodiments, the cancer is mediated by K-Ras, H-Ras and/or G12C N-Ras mutations. In other embodiments, the cancer is lung cancer, pancreatic cancer, colon cancer, MYH-associated polyposis, or colorectal cancer.
  • Route of Administration
  • The compound and the pharmaceutically acceptable salt thereof of the present invention can be prepared into various preparations which include the compound or the pharmaceutically acceptable salt thereof disclosed herein in a safe and effective amount range and a pharmaceutically acceptable excipient or carrier, wherein the "safe and effective amount" means that the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. The safe and effective amount of the compound is determined according to the age, condition, course of treatment and other specific conditions of a treated subject.
  • The "pharmaceutically acceptable excipient or carrier" refers to one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. "Compatible" means that the components of the composition are capable of intermixing with the compound of the present invention and with each other, without significantly diminishing the pharmaceutical efficacy of the compound. Examples of pharmaceutically acceptable excipients or carriers are cellulose and its derivatives (e.g., sodium carboxymethylcellulose, sodium ethylcellulose or cellulose acetate), gelatin, talc, solid lubricants (e.g., stearic acid or magnesium stearate), calcium sulfate, vegetable oil (e.g., soybean oil, sesame oil, peanut oil or olive oil), polyols (e.g., propylene glycol, glycerol, mannitol or sorbitol), emulsifiers (e.g., Tween®), wetting agents (e.g., sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • When the compound of the present invention is administered, it may be administered orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously) or topically.
  • Solid dosage forms for oral administration include capsules, tablets, pills, pulvises and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, such as starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, such as hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, such as glycerol; (d) disintegrants, such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate; (e) solution retarders, such as paraffin; (f) absorption accelerators, such as quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, such as kaolin; and (i) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycol and sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also include buffers.
  • Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may include opacifying agents, and the active compound or compound in such a composition may be released in a certain part of the digestive tract in a delayed manner. Examples of embedding components that can be used are polymeric substances and wax-based substances. If necessary, the active compound can also be in microcapsule form with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active compound, the liquid dosage form may include inert diluents commonly used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butanediol, dimethylformamide, and oils, especially cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, or mixtures of these substances. Besides such inert diluents, the composition may also include adjuvants, such as wetting agents, emulsifiers, suspending agents, sweeteners, flavoring agents, and perfuming agents. Suspensions, in addition to the active compound, may include suspending agents, such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methylate and agar, or mixtures of these substances.
  • Compositions for parenteral injection may include physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for redissolving into sterile injectable solutions or dispersions. Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof. Dosage forms for topical administration of the compound of the present invention include ointments, pulvises, patches, sprays and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers or propellants that may be required if necessary.
  • The compound of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
  • When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is administered to a mammal (such as a human) to be treated, wherein the administration dose is a pharmaceutically effective administration dose. For a human weighing 60 kg, the daily dose of administration is usually 1-2000 mg, preferably 50-1000 mg. In determining a specific dose, such factors as the route of administration, the health condition of the patient and the like will also be considered, which are well known to skilled physicians.
  • The above features mentioned in the present invention or those mentioned in the examples may be combined arbitrarily. All the features disclosed in this specification may be used with any composition form and the various features disclosed in this specification may be replaced with any alternative features that provide the same, equivalent or similar purpose. Thus, unless otherwise expressly stated, the features disclosed are merely general examples of equivalent or similar features.
  • Various specific aspects, features and advantages of the compounds, methods and pharmaceutical compositions described above are set forth in detail in the following description, which makes the present invention clear. It should be understood that the detailed description and examples below describe specific embodiments for reference only. After reading the description of the present invention, those skilled in the art can make various changes or modifications to the present invention, and such equivalents also fall within the scope of the present invention defined herein.
  • In all examples, 1H-NMR spectra were recorded with a Vian Mercury 400 nuclear magnetic resonance spectrometer, and chemical shifts are expressed in δ (ppm); silica gel for separation was 200-300 mesh silica gel if not specified, and the ratio of the eluents was volume ratio. The present invention uses the following abbreviations: CD3OD for deuterated methanol; MeCN for acetonitrile; DCM for dichloromethane; DIPEA for diisopropylethylamine; dioxane for 1,4-dioxane; DMF for dimethylformamide; K3PO4 for potassium phosphate; min for minute; MS for mass spectroscopy; NaH for sodium hydride; NMR for nuclear magnetic resonance; Pd2(dba)3 for tris(dibenzylideneacetone)dipalladium; Pd(dppf)Cl2 for [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride; TFA (CF3COOH) for trifluoroacetic acid; TLC for thin layer chromatography; THF for tetrahydrofuran; and Xantphos for 4,5-bis(diphenylphosphane)-9,9-dimethylxanthene.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1 shows the inhibition of the phosphorylated ERK (pERK) level in cells by compounds.
  • DETAILED DESCRIPTION Example 1: Synthesis of 1-(7-(6-Cyclopropyl-8-ethoxy-2-((1-(2-methoxyethyl)piperidin-4-yl)oxy)-7-(5-methyl-1H-indazol-4-yl)quinazolin-4-yl)-2,7-diazaspiro[3.5]nonan-2-yl)-2-fluoroprop-2-en-1-one (Compound 1)
  • Figure imgb0368
  • Step 1: Synthesis of compound 1-3
  • Compound 1-1 (5.5 g, 13.1 mmol) was suspended in dioxane (80 mL). DIPEA (10.1 g, 78.6 mmol) was added under ice bath, followed by 1-2 (3.0 g, 13.1 mmol). The mixture was stirred for 30 min, and stirred at room temperature for 1 h. The reaction was completed as detected by TLC. Water was added, followed by EA for extraction. The organic phase was dried and concentrated, and the residue was slurried with EA to obtain a yellow solid 1-3 (4.5 g, 56% yield).
  • 1H NMR (400 MHz, DMSO-d6 ) δ: 8.26 (d, J = 1.5 Hz, 1H), 3.79 (s, 4H), 3.65 (s, 4H), 1.86 (t, J = 5.3 Hz, 4H), 1.39 (s, 9H); MS(ESI): MS (ESI): 611.2 [M+1]+.
  • Step 2: Synthesis of compound 1-4
  • Compound 1-3 (4.5 g, 7.4 mmol) was dissolved in a mixed solution of DMF (40 mL) and THF (40 mL). 1-(2-Methoxyethyl)-4-hydroxypiperidine (2.4 g, 14.8 mmol) and DABCO (0.2 g, 1.5 mmol) were added. The mixture was stirred at room temperature overnight. After the reaction was completed, water was added, followed by EA for extraction. The organic phase was dried and concentrated, and the residue was subjected to column chromatography to obtain compound 1-4 (4.1 g, 76% yield).
  • 1H NMR (400 MHz, DMSO-d6 ) δ: 8.14 (s, 1H), 4.99 (ddd, J = 11.7, 8.5, 3.6 Hz, 1H), 3.66 (s, 9H), 3.44 (t, J = 5.8 Hz, 2H), 3.24 (s, 3H), 3.17 (d, J = 5.2 Hz, 1H), 2.77 (dt, J = 9.5, 8.8 Hz, 2H), 2.26 (t, J = 9.8 Hz, 2H), 2.00 (d, J = 12.0 Hz, 2H), 1.84 (d, J = 4.3 Hz, 4H), 1.74 - 1.61 (m, 2H), 1.39 (s, 9H); MS(ESI): 734 .2 [M+1]+.
  • Step 3: Synthesis of compound 1-5
  • Trifluoroethanol (0.9 g, 8.4 mmol) was dissolved in anhydrous DMF (10 mL). NaH was added under ice bath. The mixture was stirred at room temperature for 5 min to obtain sodium trifluoroethoxide. Compound 1-4 (4.1 g, 5.6 mmol) was dissolved in anhydrous THF (40 mL). The solution of sodium trifluoroethoxide in DMF prepared above was added. The mixture was stirred at room temperature overnight. After the reaction was completed, water was added, followed by EA for extraction. The organic phase was dried and concentrated, and the residue was subjected to column chromatography to obtain compound 1-5 (4.5 g, 99% yield). MS (ESI): 814.2 [M+1]+.
  • Step 4: Synthesis of compound 1-6
  • To a single-necked flask were added compound 1-5 (4.1 g, 5.5 mmol), cyclopropylboronic acid (0.5 g, 6.1 mmol), Pd(dppf)Cl2 (0.9 g, 1.1 mmol) and K3PO4 (0.4 g, 1.7 mmol), followed sequentially by MeCN (40 mL), dioxane (40 mL) and H2O (16.5 mL). The mixture was stirred under nitrogen at 100 °C for 5 h. After the reaction was completed, the mixture was subjected to column chromatography to obtain compound 1-6 (2.5 g, 62% yield). MS (ESI): 728.3 [M+1]+.
  • Step 5: Synthesis of compound 1-7
  • To a single-necked flask were added compound 1-6 (2.5 g, 3.4 mmol), 5-methyl-1H-indazole-4-boronic acid (0.9 g, 5.1 mmol), Pd2(dba)3 (0.3 g, 0.4 mmol), Xatphos (0.3 g, 0.7 mmol) and K3PO4 (2.2 g, 10.2 mmol), followed by dioxane (40 mL) and H2O (4 mL). The mixture was stirred under nitrogen at 120 °C overnight. After the reaction was completed, the mixture was subjected to column chromatography to obtain compound 1-7 (1 g, 38% yield). MS (ESI): 780.4 [M+1]+.
  • Step 6: Synthesis of compound 1-8
  • Compound 1-7 (1 g, 1.3 mmol) was dissolved in DCM (15 mL). TFA (5 mL) was added. The mixture was stirred at room temperature for 2 h. After the reaction was completed, the mixture was concentrated, basified with saturated sodium carbonate, and extracted with EA. The organic phase was dried and concentrated to obtain compound 1-8 (0.9 g, 99% yield). MS(ESI): 680.4 [M+1]+.
  • Step 7: Synthesis of compound 1
  • Compound 1-8 (150 mg, 0.2 mmol) and 2-fluoroacrylic acid (20 mg, 0.22 mmol) were dissolved in DCM (15 mL). DIPEA (52 mg, 0.4 mmol) and HATU (114 mg, 0.3 mmol) were added under ice bath. The mixture was stirred overnight. After the reaction was completed, the reaction mixture was washed with saturated brine. The organic phase was dried and concentrated, and the residue was subjected to column chromatography to obtain compound 1 (30 mg, 20% yield).
  • 1H NMR (400 MHz, CD3OD) δ: 7.47-7.37 (m, 2H), 7.30 (d, J = 8.6 Hz, 1H), 7.24 (s, 1H), 5.52 (d, J= 3.4 Hz, 0.5H), 5.40 (d, J= 3.4 Hz, 0.5H), 5.15 (d, J= 3.4 Hz, 0.5H), 5.11 (d, J= 3.4 Hz, 0.5H), 4.54 (dq, J = 17.7, 8.8 Hz, 1H), 4.23-4.12 (m, 3H), 3.83 (s, 2H), 3.73-3.59 (m, 4H), 3.54 (t, J = 5.2 Hz, 2H), 3.28 (s, 3H), 3.10 (dd, J = 10.1, 6.2 Hz, 2H), 2.88 (s, 2H), 2.85-2.72 (m, 2H), 2.13 (d, J = 26.2 Hz, 5H), 1.97 (dd, J = 11.9, 6.8 Hz, 6H), 1.36 (dt, J = 14.0, 6.6 Hz, 1H), 1.27-1.17 (m, 5H); MS (ESI): 752.4 [M+1]+.
  • Example 2-341: Synthesis of Compound 2-341
  • The target compound 2-341 was obtained using different starting materials according to a synthesis method similar to that in Example 1. Table 1
    Compound Compound structure [M+H]+ Compound Compound structure [M+H]+
    2
    Figure imgb0369
    698.4 3
    Figure imgb0370
    734.4
    4
    Figure imgb0371
    684.4 5
    Figure imgb0372
    738.3
    6
    Figure imgb0373
    792.4 7
    Figure imgb0374
    747.4
    8
    Figure imgb0375
    691.4 9
    Figure imgb0376
    727.4
    10
    Figure imgb0377
    694.3 11
    Figure imgb0378
    712.3
    12
    Figure imgb0379
    728.3 13
    Figure imgb0380
    708.3
    14
    Figure imgb0381
    722.4 15
    Figure imgb0382
    736.4
    16
    Figure imgb0383
    718.3 17
    Figure imgb0384
    748.4
    18
    Figure imgb0385
    759.3 19
    Figure imgb0386
    802.3
    20
    Figure imgb0387
    812.4 21
    Figure imgb0388
    812.4
    22
    Figure imgb0389
    800.4 23
    Figure imgb0390
    801.4
    24
    Figure imgb0391
    799.4 25
    Figure imgb0392
    801.4
    26
    Figure imgb0393
    706.3 27
    Figure imgb0394
    734.4
    28
    Figure imgb0395
    767.4 29
    Figure imgb0396
    786.3
    30
    Figure imgb0397
    770.4 31
    Figure imgb0398
    766.4
    32
    Figure imgb0399
    752.3 33
    Figure imgb0400
    756.3
    34
    Figure imgb0401
    756.3 35
    Figure imgb0402
    734.4
    36
    Figure imgb0403
    756.3 37
    Figure imgb0404
    738.3
    38
    Figure imgb0405
    756.3 39
    Figure imgb0406
    735.4
    40
    Figure imgb0407
    753.3 41
    Figure imgb0408
    761.3
    42
    Figure imgb0409
    779.3 43
    Figure imgb0410
    762.3
    44
    Figure imgb0411
    780.3 45
    Figure imgb0412
    714.3
    46
    Figure imgb0413
    732.3 47
    Figure imgb0414
    713.4
    48
    Figure imgb0415
    731.3 49
    Figure imgb0416
    781.3
    50
    Figure imgb0417
    799.2 51
    Figure imgb0418
    698.3
    52
    Figure imgb0419
    715.3 53
    Figure imgb0420
    716.3
    54
    Figure imgb0421
    734.3 55
    Figure imgb0422
    748.3
    56
    Figure imgb0423
    715.3 57
    Figure imgb0424
    714.3
    58
    Figure imgb0425
    730.3 59
    Figure imgb0426
    746.3
    60
    Figure imgb0427
    746.3 61
    Figure imgb0428
    708.3
    62
    Figure imgb0429
    750.3 63
    Figure imgb0430
    766.4
    64
    Figure imgb0431
    694.3 65
    Figure imgb0432
    738.3
    66
    Figure imgb0433
    736.3 67
    Figure imgb0434
    674.3
    68
    Figure imgb0435
    692.3 69
    Figure imgb0436
    687.3
    70
    Figure imgb0437
    705.3 71
    Figure imgb0438
    731.4
    72
    Figure imgb0439
    749.3 73
    Figure imgb0440
    701.3
    74
    Figure imgb0441
    719.3 75
    Figure imgb0442
    745.4
    76
    Figure imgb0443
    763.4 77
    Figure imgb0444
    715.4
    78
    Figure imgb0445
    733.4 79
    Figure imgb0446
    741.4
    80
    Figure imgb0447
    759.4 81
    Figure imgb0448
    771.4
    82
    Figure imgb0449
    789.4 83
    Figure imgb0450
    755.4
    84
    Figure imgb0451
    773.4 85
    Figure imgb0452
    783.3
    86
    Figure imgb0453
    801.3 87
    Figure imgb0454
    754.4
    88
    Figure imgb0455
    772.4 89
    Figure imgb0456
    757.4
    90
    Figure imgb0457
    775.4 91
    Figure imgb0458
    759.4
    92
    Figure imgb0459
    777.4 93
    Figure imgb0460
    773.4
    94
    Figure imgb0461
    791.4 95
    Figure imgb0462
    773.4
    96
    Figure imgb0463
    791.4 97
    Figure imgb0464
    701.3
    98
    Figure imgb0465
    719.3 99
    Figure imgb0466
    745.4
    100
    Figure imgb0467
    763.4 101
    Figure imgb0468
    773.4
    102
    Figure imgb0469
    791.4 103
    Figure imgb0470
    759.4
    104
    Figure imgb0471
    777.4 105
    Figure imgb0472
    773.4
    106
    Figure imgb0473
    791.4 107
    Figure imgb0474
    787.4
    108
    Figure imgb0475
    805.4 109
    Figure imgb0476
    662.3
    110
    Figure imgb0477
    680.3 111
    Figure imgb0478
    704.3
    112
    Figure imgb0479
    722.3 113
    Figure imgb0480
    706.3
    114
    Figure imgb0481
    724.3 115
    Figure imgb0482
    706.3
    116
    Figure imgb0483
    723.3 117
    Figure imgb0484
    750.4
    118
    Figure imgb0485
    768.3 119
    Figure imgb0486
    746.4
    120
    Figure imgb0487
    764.4 121
    Figure imgb0488
    746.4
    122
    Figure imgb0489
    764.4 123
    Figure imgb0490
    702.3
    124
    Figure imgb0491
    720.3 125
    Figure imgb0492
    746.3
    126
    Figure imgb0493
    764.3 127
    Figure imgb0494
    744.3
    128
    Figure imgb0495
    762.3 129
    Figure imgb0496
    730.4
    130
    Figure imgb0497
    748.4 131
    Figure imgb0498
    774.4
    132
    Figure imgb0499
    792.4 133
    Figure imgb0500
    772.4
    134
    Figure imgb0501
    790.4 135
    Figure imgb0502
    730.4
    136
    Figure imgb0503
    748.4 137
    Figure imgb0504
    773.4
    138
    Figure imgb0505
    792.4 139
    Figure imgb0506
    693.3
    140
    Figure imgb0507
    719.4 141
    Figure imgb0508
    737.3
    142
    Figure imgb0509
    687.3 143
    Figure imgb0510
    705.3
    144
    Figure imgb0511
    717.3 145
    Figure imgb0512
    735.3
    146
    Figure imgb0513
    717.3 147
    Figure imgb0514
    735.3
    148
    Figure imgb0515
    745.4 149
    Figure imgb0516
    763.4
    150
    Figure imgb0517
    702.3 151
    Figure imgb0518
    720.3
    152
    Figure imgb0519
    717.3 153
    Figure imgb0520
    735.3
    154
    Figure imgb0521
    745.4 155
    Figure imgb0522
    763.4
    156
    Figure imgb0523
    702.3 157
    Figure imgb0524
    727.4
    158
    Figure imgb0525
    729.3 159
    Figure imgb0526
    715.4
    160
    Figure imgb0527
    759.4 161
    Figure imgb0528
    729.4
    162
    Figure imgb0529
    715.4 163
    Figure imgb0530
    729.4
    164
    Figure imgb0531
    743.4 165
    Figure imgb0532
    689.3
    166
    Figure imgb0533
    703.4 167
    Figure imgb0534
    743.3
    168
    Figure imgb0535
    705.3 169
    Figure imgb0536
    723.3
    170
    Figure imgb0537
    731.4 171
    Figure imgb0538
    700.4
    172
    Figure imgb0539
    719.3 173
    Figure imgb0540
    719.3
    174
    Figure imgb0541
    731.4 175
    Figure imgb0542
    731.4
    176
    Figure imgb0543
    737.3 177
    Figure imgb0544
    715.4
    178
    Figure imgb0545
    733.3 179
    Figure imgb0546
    745.4
    180
    Figure imgb0547
    751.3 181
    Figure imgb0548
    700.4
    182
    Figure imgb0549
    700.4 183
    Figure imgb0550
    715.4
    184
    Figure imgb0551
    715.4 185
    Figure imgb0552
    689.4
    186
    Figure imgb0553
    689.4 187
    Figure imgb0554
    703.4
    188
    Figure imgb0555
    703.4 189
    Figure imgb0556
    717.4
    190
    Figure imgb0557
    717.4 191
    Figure imgb0558
    733.4
    192
    Figure imgb0559
    733.4 193
    Figure imgb0560
    733.4
    194
    Figure imgb0561
    733.4 195
    Figure imgb0562
    745.4
    196
    Figure imgb0563
    745.4 197
    Figure imgb0564
    745.4
    198
    Figure imgb0565
    745.4 199
    Figure imgb0566
    751.3
    200
    Figure imgb0567
    751.3 201
    Figure imgb0568
    719.3
    202
    Figure imgb0569
    719.3 203
    Figure imgb0570
    731.4
    204
    Figure imgb0571
    731.3 205
    Figure imgb0572
    737.3
    206
    Figure imgb0573
    737.3 207
    Figure imgb0574
    729.4
    208
    Figure imgb0575
    729.4 209
    Figure imgb0576
    731.4
    210
    Figure imgb0577
    731.4 211
    Figure imgb0578
    747.4
    212
    Figure imgb0579
    747.4 213
    Figure imgb0580
    759.4
    214
    Figure imgb0581
    759.4 215
    Figure imgb0582
    715.4
    216
    Figure imgb0583
    729.4 217
    Figure imgb0584
    701.4
    218
    Figure imgb0585
    715.4 219
    Figure imgb0586
    705.3
    220
    Figure imgb0587
    719.3 221
    Figure imgb0588
    687.3
    222
    Figure imgb0589
    701.4 223
    Figure imgb0590
    691.3
    224
    Figure imgb0591
    705.3 225
    Figure imgb0592
    677.4
    226
    Figure imgb0593
    688.4 227
    Figure imgb0594
    703.4
    228
    Figure imgb0595
    707.3 229
    Figure imgb0596
    705.3
    230
    Figure imgb0597
    719.4 231
    Figure imgb0598
    714.3
    232
    Figure imgb0599
    703.4 233
    Figure imgb0600
    717.4
    234
    Figure imgb0601
    719.4 235
    Figure imgb0602
    733.4
    236
    Figure imgb0603
    721.4 237
    Figure imgb0604
    701.4
    238
    Figure imgb0605
    712.3 239
    Figure imgb0606
    785.3
    240
    Figure imgb0607
    717.3 241
    Figure imgb0608
    765.3
    242
    Figure imgb0609
    726.3 243
    Figure imgb0610
    717.3
    244
    Figure imgb0611
    730.4 245
    Figure imgb0612
    715.4
    246
    Figure imgb0613
    715.4 247
    Figure imgb0614
    715.4
    248
    Figure imgb0615
    715.4 249
    Figure imgb0616
    726.3
    250
    Figure imgb0617
    726.3 251
    Figure imgb0618
    731.4
    252
    Figure imgb0619
    731.4 253
    Figure imgb0620
    759.4
    254
    Figure imgb0621
    759.4 255
    Figure imgb0622
    744.4
    256
    Figure imgb0623
    744.4 257
    Figure imgb0624
    726.3
    258
    Figure imgb0625
    726.3 259
    Figure imgb0626
    731.4
    260-
    Figure imgb0627
    731.4 261
    Figure imgb0628
    735.3
    262
    Figure imgb0629
    735.3 263
    Figure imgb0630
    749.4
    264
    Figure imgb0631
    749.4 265
    Figure imgb0632
    731.4
    266
    Figure imgb0633
    745.4 267
    Figure imgb0634
    737.3
    268
    Figure imgb0635
    729.4 269
    Figure imgb0636
    743.4
    270
    Figure imgb0637
    727.4 271
    Figure imgb0638
    717.3
    272
    Figure imgb0639
    717.3 273
    Figure imgb0640
    719.3
    274
    Figure imgb0641
    719.3 275
    Figure imgb0642
    731.4
    276
    Figure imgb0643
    731.4 277
    Figure imgb0644
    744.4
    278
    Figure imgb0645
    744.4 279
    Figure imgb0646
    688.3
    280
    Figure imgb0647
    731.4 281
    Figure imgb0648
    731.4
    282
    Figure imgb0649
    688.3 283
    Figure imgb0650
    702.3
    284
    Figure imgb0651
    690.3 285
    Figure imgb0652
    697.3
    286
    Figure imgb0653
    715.4 287
    Figure imgb0654
    727.4
    288
    Figure imgb0655
    741.4 289
    Figure imgb0656
    741.4
    290
    Figure imgb0657
    755.4 291
    Figure imgb0658
    755.4
    292
    Figure imgb0659
    755.4 293
    Figure imgb0660
    755.4
    294
    Figure imgb0661
    729.4 295
    Figure imgb0662
    729.4
    296
    Figure imgb0663
    715.4 297
    Figure imgb0664
    715.4
    298
    Figure imgb0665
    729.4 299
    Figure imgb0666
    729.4
    300
    Figure imgb0667
    729.4 301
    Figure imgb0668
    729.4
    302
    Figure imgb0669
    741.4 303
    Figure imgb0670
    741.4
    304
    Figure imgb0671
    690.3 305
    Figure imgb0672
    706.3
    306
    Figure imgb0673
    721.3 307
    Figure imgb0674
    750.3
    308
    Figure imgb0675
    702.3 309
    Figure imgb0676
    746.3
    310
    Figure imgb0677
    751.3 311
    Figure imgb0678
    753.3
    312
    Figure imgb0679
    751.3 313
    Figure imgb0680
    751.3
    314
    Figure imgb0681
    737.3 315
    Figure imgb0682
    737.3
    316
    Figure imgb0683
    739.3 317
    Figure imgb0684
    753.3
    318
    Figure imgb0685
    768.3 319
    Figure imgb0686
    768.3
    320
    Figure imgb0687
    687.3 321
    Figure imgb0688
    701.3
    322
    Figure imgb0689
    701.3 323
    Figure imgb0690
    715.3
    324
    Figure imgb0691
    729.3 325
    Figure imgb0692
    729.3
    326
    Figure imgb0693
    741.4 327
    Figure imgb0694
    733.4
    328
    Figure imgb0695
    733.4 329
    Figure imgb0696
    745.4
    330
    Figure imgb0697
    661.4 331
    Figure imgb0698
    661.4
    332
    Figure imgb0699
    673.4 333
    Figure imgb0700
    697.4
    334
    Figure imgb0701
    697.4 335
    Figure imgb0702
    709.4
    336
    Figure imgb0703
    673.3 337
    Figure imgb0704
    687.4
    338
    Figure imgb0705
    745.4 339
    Figure imgb0706
    701.4
    340
    Figure imgb0707
    701.4 341
    Figure imgb0708
    713.4
  • Example 342: Chiral Resolution of Compound 142
  • The compounds of the present application may have axial chirality. Compounds with axial chirality can be resolved to obtain two chiral isomers.
  • Compound 142 (50 mg) was dissolved in ethanol (2 mL) at a concentration of 25 mg/mL. The volume for each injection was 500 µL. Conditions for preparative chromatography: CHIRALPAK AD-H (20 × 250 mm, 5 µm) chromatography column; mobile phase: ethanol-n-hexane (40/60); flow rate: 12 mL/min; wavelength of detection: 254 nm. The stepwise eluate was concentrated by rotary evaporation and dried to obtain two chiral isomers 142-a and 142-b of compound 142:
    • a first chiral isomer: 142-a; retention time on the chromatography column: 6.662 min; and
    • a second chiral isomer: 142-b; retention time on the chromatography column: 10.831 min.
  • Compounds 171, 174 and 270 were chirally resolved using a similar resolution procedure to obtain their two chiral isomers 171-a/171-b, 174-a/174-b and 270-a/270-b, respectively. Their retention times on the chromatography column are as follows: Table 2. Conditions for and results of chiral resolution of compounds 171, 174 and 270
    Compound Conditions for resolution Results of resolution
    171 Same as those for compound 142 a first chiral isomer: 171-a; retention time on the chromatography column: 7.481 min; and a second chiral isomer: 171-b; retention time on the chromatography column: 12.770 min.
    174 Same as those for compound 142 a first chiral isomer: 174-a; retention time on the chromatography column: 8.994 min; and a second chiral isomer: 174-b; retention time on the chromatography column: 14.583 min.
    270 Same as those for compound 142 except that the mobile phase is ethanol-n-hexane (30/70) a first chiral isomer: 270-a; retention time on the chromatography column: 7.280 min; and a second chiral isomer: 270-b; retention time on the chromatography column: 12.962 min.
  • Other compounds in the present application can also be chirally resolved using a similar method.
  • Example 343: pERK and ERK Protein Content Assay in H358 Cells by Compounds
  • H358 cells were seeded in a 24-well plate. After one day of growth, a test compound (at a concentration of 1 µM) was added. After 24 h of action of the compound, the cells were lysed, and the cell lysate was transferred to a 96-well ELISA plate. The levels of pERK and ERK in the lysate were measured using an ELISA kit (abcam 176660). The ratio of pERK to ERK was calculated and compared with that of the DMSO group, and the percentage of inhibition of pERK activity by the compound was calculated. The results are shown in Table 3 below. Table 3. Inhibitory activity of the compounds of the present invention against the pERK level in H358 cells
    Compound Inhibition rate (%) Compound Inhibition rate (%) Compound Inhibition rate (%)
    1 +++ 2 +++ 3 +++
    4 +++ 5 +++ 6 +++
    7 ++ 8 ++ 9 +++
    10 ++ 11 +++ 12 +++
    13 +++ 14 +++ 15 +++
    16 ++ 17 +++ 18 ++
    19 ++ 20 +++ 21 +++
    22 ++ 23 +++ 24 ++
    25 +++ 26 ++ 27 ++
    28 ++ 29 +++ 30 +++
    31 +++ 32 +++ 33 +++
    34 +++ 35 +++ 36 +++
    37 ++ 38 +++ 39 +++
    40 +++ 41 +++ 42 +++
    43 +++ 44 +++ 45 +++
    46 +++ 47 +++ 48 +++
    49 +++ 50 +++ 51 +++
    52 +++ 53 +++ 54 +++
    55 +++ 56 +++ 57 +++
    58 +++ 59 +++ 60 +++
    61 +++ 62 +++ 63 +++
    64 ++ 65 +++ 66 ++
    67 ++ 68 ++ 69 +++
    70 +++ 71 +++ 72 +++
    73 +++ 74 +++ 75 +++
    76 +++ 77 ++ 78 ++
    79 ++ 80 ++ 81 +++
    82 +++ 83 +++ 84 +++
    85 +++ 86 ++ 87 +++
    88 ++ 89 +++ 90 +++
    91 +++ 92 +++ 93 +++
    94 +++ 95 +++ 96 +++
    97 ++ 98 ++ 99 +++
    100 +++ 101 +++ 102 +++
    103 +++ 104 ++ 105 +++
    106 ++ 107 ++ 108 ++
    109 +++ 110 +++ 111 +++
    112 ++ 113 +++ 114 +++
    115 ++ 116 ++ 117 +++
    118 +++ 119 ++ 120 ++
    121 ++ 122 ++ 123 +++
    124 +++ 125 +++ 126 +++
    127 +++ 128 +++ 129 +++
    130 +++ 131 +++ 132 +++
    133 +++ 134 +++ 135 ++
    136 ++ 137 +++ 138 +++
    139 +++ 140 +++ 141 +++
    142 +++ 143 +++ 144 +++
    145 +++ 146 +++ 147 +++
    148 ++ 149 ++ 150 +++
    151 +++ 152 +++ 153 +++
    154 +++ 155 ++ 156 +++
    157 +++ 158 +++ 159 +++
    160 +++ 161 +++ 162 +++
    163 +++ 164 +++ 165 +++
    166 +++ 167 +++ 168 +++
    169 ++ 170 ++ 171 +++
    172 +++ 173 +++ 174 +++
    175 +++ 176 ++ 177 +++
    178 +++ 179 +++ 180 +++
    181 +++ 182 +++ 183 +++
    184 +++ 185 +++ 186 +++
    187 +++ 188 +++ 189 +++
    190 +++ 191 +++ 192 +++
    193 +++ 194 +++ 195 +++
    196 +++ 197 +++ 198 +++
    199 +++ 200 +++ 201 +++
    202 +++ 203 +++ 204 +++
    205 ++ 206 ++ 207 +++
    208 +++ 209 +++ 210 +++
    211 +++ 212 +++ 213 ++
    214 ++ 215 +++ 216 +++
    217 +++ 218 +++ 219 +++
    220 +++ 221 +++ 222 +++
    223 +++ 224 +++ 225 +++
    226 +++ 227 +++ 228 +++
    229 +++ 230 ++ 231 ++
    232 +++ 233 +++ 234 +++
    235 +++ 236 +++ 237 +++
    238 +++ 239 ++ 240 +++
    241 ++ 242 +++ 243 +++
    244 +++ 245 +++ 246 +++
    247 +++ 248 +++ 249 ++
    250 ++ 251 +++ 252 +++
    253 +++ 254 +++ 255 +++
    256 +++ 257 +++ 258 +++
    259 +++ 260 +++ 261 +++
    262 +++ 263 +++ 264 +++
    265 +++ 266 +++ 267 +++
    268 +++ 269 +++ 270 +++
    271 +++ 272 +++ 273 +++
    274 +++ 275 +++ 276 +++
    277 +++ 278 +++ 279 +++
    280 +++ 281 +++ 282 +++
    283 ++ 284 +++ 285 ++
    286 +++ 287 +++ 288 +++
    289 +++ 290 +++ 291 +++
    292 +++ 293 +++ 294 +++
    295 +++ 296 +++ 297 +++
    298 +++ 299 +++ 300 +++
    301 +++ 302 +++ 303 +++
    304 ++ 305 ++ 306 +++
    307 +++ 308 +++ 309 +++
    310 ++ 311 +++ 312 +++
    313 +++ 314 +++ 315 +++
    316 +++ 317 +++ 318 +++
    319 +++ 320 +++ 321 +++
    322 +++ 323 +++ 324 +++
    325 +++ 326 +++ 327 +++
    328 +++ 329 +++ 330 +++
    331 +++ 332 +++ 333 +++
    334 +++ 335 +++ 336 +++
    337 +++ 338 +++ 339 +++
    340 +++ 341 +++ 142-a +++
    142-b ++ 171-a +++ 171-b +++
    174-a +++ 174-b ++ 270-a +++
    270-b +++ B +++ C +++
    + indicates an inhibition rate less than or equal to 50%
    ++ indicates an inhibition rate from 50% to 90%
    +++ indicates an inhibition rate greater than 90%.
  • Example 344: Antiproliferative Activity of Compounds Against H358 cells
  • 2500 H358 cells were seeded in a 96-well ultra-low attachment plate (corning, 7007). After one day of growth, a serially diluted compound (a maximum concentration of 5 µM, 5-fold dilution, a total of five doses) was added. Three days after the addition of the compound, Cell Titer Glow (Promega, G9681) was added to evaluate pellet growth, and the IC50 value was calculated. The results are shown in Table 4 below. Table 4. Antiproliferative activity of the compounds of the present invention against H358 cells
    Compound IC50 Compound IC50 Compound IC50
    1 +++ 2 +++ 3 +++
    4 +++ 5 +++ 6 +++
    7 ++ 8 ++ 9 +++
    10 ++ 11 +++ 12 +++
    13 ++ 14 ++ 15 +++
    16 ++ 17 +++ 18 ++
    19 ++ 20 +++ 21 +++
    22 ++ 23 +++ 24 ++
    25 +++ 26 ++ 27 ++
    28 ++ 29 +++ 30 +++
    31 +++ 32 +++ 33 +++
    34 +++ 35 +++ 36 +++
    37 ++ 38 ++ 39 +++
    40 +++ 41 +++ 42 ++
    43 +++ 44 +++ 45 +++
    46 +++ 47 +++ 48 ++
    49 +++ 50 +++ 51 +++
    52 +++ 53 +++ 54 +++
    55 +++ 56 +++ 57 +++
    58 +++ 59 +++ 60 +++
    61 +++ 62 +++ 63 +++
    64 ++ 65 +++ 66 ++
    67 ++ 68 ++ 69 +++
    70 ++ 71 +++ 72 +++
    73 +++ 74 +++ 75 +++
    76 +++ 77 ++ 78 ++
    79 ++ 80 ++ 81 +++
    82 +++ 83 +++ 84 +++
    85 ++ 86 ++ 87 +++
    88 ++ 89 +++ 90 +++
    91 +++ 92 +++ 93 +++
    94 +++ 95 +++ 96 +++
    97 ++ 98 ++ 99 +++
    100 +++ 101 +++ 102 +++
    103 +++ 104 ++ 105 +++
    106 ++ 107 ++ 108 ++
    109 +++ 110 +++ 111 +++
    112 ++ 113 +++ 114 +++
    115 ++ 116 ++ 117 +++
    118 +++ 119 +++ 120 ++
    121 +++ 122 ++ 123 +++
    124 +++ 125 +++ 126 +++
    127 +++ 128 +++ 129 +++
    130 +++ 131 +++ 132 +++
    133 +++ 134 +++ 135 ++
    136 ++ 137 +++ 138 +++
    139 +++ 140 +++ 141 +++
    142 +++ 143 +++ 144 +++
    145 +++ 146 +++ 147 +++
    148 ++ 149 ++ 150 +++
    151 ++ 152 +++ 153 +++
    154 +++ 155 ++ 156 +++
    157 +++ 158 +++ 159 +++
    160 +++ 161 +++ 162 +++
    163 +++ 164 +++ 165 +++
    166 +++ 167 ++ 168 +++
    169 ++ 170 ++ 171 +++
    172 +++ 173 +++ 174 +++
    175 +++ 176 ++ 177 +++
    178 +++ 179 +++ 180 ++
    181 +++ 182 +++ 183 +++
    184 +++ 185 +++ 186 +++
    187 +++ 188 +++ 189 +++
    190 +++ 191 +++ 192 +++
    193 +++ 194 +++ 195 +++
    196 +++ 197 +++ 198 +++
    199 ++ 200 ++ 201 +++
    202 +++ 203 ++ 204 ++
    205 ++ 206 ++ 207 +++
    208 +++ 209 +++ 210 +++
    211 +++ 212 +++ 213 ++
    214 ++ 215 +++ 216 +++
    217 +++ 218 +++ 219 +++
    220 +++ 221 +++ 222 +++
    223 +++ 224 +++ 225 +++
    226 +++ 227 +++ 228 +++
    229 +++ 230 ++ 231 ++
    232 +++ 233 +++ 234 +++
    235 +++ 236 +++ 237 +++
    238 ++ 239 ++ 240 +++
    241 ++ 242 +++ 243 +++
    244 +++ 245 +++ 246 +++
    247 +++ 248 +++ 249 ++
    250 ++ 251 +++ 252 +++
    253 ++ 254 ++ 255 +++
    256 +++ 257 ++ 258 ++
    259 +++ 260 +++ 261 +++
    262 +++ 263 +++ 264 +++
    265 +++ 266 +++ 267 +++
    268 +++ 269 +++ 270 +++
    271 +++ 272 +++ 273 +++
    274 +++ 275 +++ 276 +++
    277 +++ 278 +++ 279 +++
    280 +++ 281 +++ 282 +++
    283 ++ 284 +++ 285 ++
    286 +++ 287 +++ 288 +++
    289 +++ 290 +++ 291 +++
    292 +++ 293 +++ 294 +++
    295 +++ 296 +++ 297 +++
    298 +++ 299 +++ 300 +++
    301 +++ 302 +++ 303 +++
    304 ++ 305 ++ 306 +++
    307 +++ 308 +++ 309 +++
    310 ++ 311 +++ 312 +++
    313 +++ 314 +++ 315 +++
    316 +++ 317 +++ 318 +++
    319 +++ 320 +++ 321 +++
    322 +++ 323 +++ 324 +++
    325 +++ 326 +++ 327 +++
    328 +++ 329 +++ 330 +++
    331 +++ 332 +++ 333 +++
    334 +++ 335 +++ 336 +++
    337 +++ 338 +++ 339 +++
    340 +++ 341 +++ 142-a +++
    142-b ++ 171-a +++ 171-b +++
    174-a +++ 174-b ++ 270-a +++
    270-b +++ B +++ C +++
    + indicates the IC50 of the compound is greater than 1 µM
    ++ indicates the IC50 of the compound is from 0.3 to 1 µM
    +++ indicates the IC50 of the compound is less than 0.3 µM.
  • As can be seen from the data in Tables 3 and 4, the antiproliferative activity of most of the compounds of the present invention against H358 cells is less than 0.3 µM, and when R5 (or R5a or R5b) is a spiro ring or other substituted heterocyclic ring, the compounds have very high K-RAS G12C inhibitory activity. Compounds 131, 142 and 171 all have good antiproliferative activity against H358 cells, with their IC50 values being 1.5 nM, 2.5 nM and 1.4 nM, respectively, while the IC50 values of the reference compounds B and C were 4.6 nM and 5.1 nM, respectively, indicating that the cell activity of the compounds was greatly improved after cyclization of the amino groups on the side chains of the compounds. In addition, when position 2 (substituent R4) of acrylamide is substituted with a F atom that is small in size, the compounds also have very high K-RAS G12C inhibitory activity.
  • Example 345: Pharmacokinetic Evaluation in Mice
  • The compounds were administered by intravenous injection at a dose of 2 mg/kg and oral gavage at a dose of 10 mg/kg (0.5% CMC-Na suspension). 15 male ICR mice were selected for each group, and each mouse was subjected to blood collection at 3 discrete time points, with 3 mice per time point. The time points of sampling were as follows: before the administration, and at 5 min, 15 min, 30 min, 1 h, 3 h, 5 h, 8 h, 12 h and 24 h after the administration. 80 µL of blood was collected from the eye sockets or the hearts of the mice at each of the time points after the administration. All whole blood samples were collected in tubes containing EDTA K2 and centrifuged (1500-1600 rmp) at 4 °C for 10 min to isolate plasma, which was then stored in a refrigerator at -90 to -60 °C for sample analysis. The compound concentration in the plasma was determined by liquid chromatography-tandem mass spectrometry, and the corresponding pharmacokinetic parameters were obtained according to a plasma concentration-time curve. Table 5. Pharmacokinetic parameters of compounds in mice
    Compound Route of administration Dose (mg/kg) t ½ (h) Tmax (h) Cmax (ng/mL) AUC 0-t (ng.h/L) Vss (mL/kg) Cl (mL/h/kg) F (%)
    1 iv 2 4.31 1 2557 13987 782 140 NA
    po
    10 6.94 4 1543 20304 NA NA 29.0
    131 iv 2 4.12 0.5 2346 11340 2104 112 NA
    po
    10 5.86 2 1824 22315 NA NA 39.4
    142 iv 2 3.31 0.083 13067 51995 187 38.5 NA
    po
    10 3.89 2 6730 45952 NA NA 17.7%
    171 iv 2 4.85 0.083 4910 26500 505 75.6 NA
    po
    10 3.93 2 3320 35700 NA NA 26.9
    B iv 2 4.47 0.083 2883 7822 1010 252 NA
    po
    10 3.74 2 1300 10348 -NA NA 26.5
    C iv 2 4.02 0.083 3210 20200 481 99 NA
    po
    10 3.38 0.5 3110 24800 NA NA 24.6
    NA indicates data are not available;
  • As can be seen from the above table, compared to compound B, compound 131 has good oral absorption properties, and has improved metabolic parameters such as half-life (t½), maximum plasma concentration (Cmax), area under the drug-time curve (AUC0-t), and oral bioavailability. It should be particularly noted that, compared to the reference compound C in the patent (Example 65 of WO2018/143315 ), compound 171 has better metabolic parameters, and compound 142 also has significantly improved metabolic parameters such as Cmax and AUC0-t, indicating that the metabolic properties of the compound are well improved after the amino groups on the side chain are cyclized. The metabolic properties of the compounds similar to compounds 131 and 171 in the present application are also significantly improved. Good oral absorption properties are of great significance in improving the efficacy of drugs, reducing the dose of administration and reducing the costs.
  • Example 346: Evaluation of Antitumor Activity in Mice
  • Human pancreatic cancer Mia PaCa-2 cells were cultured conventionally in 1640 medium containing 10% fetal bovine serum in a 37 °C/5% CO2 incubator. After passage, the cells were collected when they reached the desired amount. 1×107 Mia PaCa-2 cells were injected into the left dorsal side of each nude mouse, and the animals were randomly grouped for administration after tumors grew to 150 mm3. The groups are as follows: 1) a solvent control group of 8 mice; and 2) compound 1 group, compound 2 group, compound 5 group, compound 31 group, compound 131 group, compound 142 group, compound 171 group, compound B group and compound C group, with 8 mice per group. Mice in the solvent control group were subjected to intragastric administration of 0.5% CMC-Na once daily; mice in compound 1 group, compound 2 group, compound 5 group, compound 31 group, compound 131 group, compound 142 group, compound 171 group, compound B group and compound C group were subjected to intragastric administration of a suspension of a compound in 0.5% CMC-Na once daily. On Tuesday and Thursday each week, tumor volumes and body weight of the mice were measured, and the nude mice were sacrificed on day 21 of administration. The test results are shown in Table 6 below. Table 6. Experimental therapeutic effects of compounds on graft tumors of human pancreatic cancer Mia PaCa-2 in nude mice
    Compound Dose (mg/kg) Administration regimen Anti-tumor effect
    1 10 qd21 39% regression
    2 10 qd21 23% regression
    5 10 qd21 30% regression
    31 10 qd21 32% regression
    131 10 qd21 37% regression
    142 10 qd21 35% regression
    171 10 qd21 38% regression
    B
    10 qd21 25% regression
    C
    10 qd21 8% regression
  • As can be seen from the data in the table above, the compounds of the present invention have high in vivo antitumor activity; a tumor can regress after 21 consecutive days of administration at 10 mg/kg/day; compounds 1, 5, 31, 131, 142 and 171 have higher in vivo activity than reference compound B and compound C, and compounds 142 and 171 have significantly higher in vivo activity than compound C, indicating the in vivo activity of the compound is also greatly improved after the amino groups on the side chain of the compound are cyclized.
  • Example 346: pERK Level Assay by Western Blot
  • H358 cells were plated on to a 24-well plate at 2 × 105 cells/well. Serially diluted compounds including AMG510, MRTX849, compound 142 and compound 171 were added. After overnight incubation, cells were lysed, and proteins were quantified and subjected to gel electrophoresis.
  • The results of the phosphorylated ERK (pERK) level assay by western blot are shown in FIG. 1.
  • As can be seen from the results in FIG. 1, the compounds 142 and 171 of the present invention shows stronger inhibition of the phosphorylated ERK (pERK) level in cells than the reference drugs AMG510 and MRTX849 when at the same concentration.

Claims (17)

  1. A compound with a structure as shown in general formula (1), isomers thereof, crystalline forms thereof, pharmaceutically acceptable salts thereof, hydrates thereof or solvates thereof:
    Figure imgb0709
    wherein in formula (1):
    R1 is H, halogen, C1-C3 alkyl, C2-C4 alkenyl, C2-C4 alkynyl or C3-C6 cycloalkyl;
    R2 is C1-C3 alkoxy, C1-C3 haloalkoxy or -NRaRb, wherein Ra and Rb are independently H, Cl-C3 alkyl or C1-C3 haloalkyl, or Ra and Rb, together with a N atom, form a 4-7 membered heterocycloalkyl group, wherein the heterocycloalkyl group may be substituted with 1-3 halogen atoms;
    R3 is
    Figure imgb0710
    Figure imgb0711
    Figure imgb0712
    wherein Rc is H or F; Rdis H, F, Cl or Me; Re is H, F, Cl or Me; Rf is F, NH2, Me or cyclopropyl; Rx1, Rx2, Rx3, Rx4, Rx5, Rx6 and Rx7 are independently H, F, Cl, OH, OMe, NH2, CF3, C1-C3 alkyl or C3-C6 cycloalkyl;
    R4 is H, halogen, CN, C1-C3 alkyl, C1-C3 haloalkyl or heteroaryl; and
    when R3 is
    Figure imgb0713
    and R4 is H, R5is:
    Figure imgb0714
    Figure imgb0715
    Figure imgb0716
    Figure imgb0717
    Figure imgb0718
    wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rh is
    Figure imgb0719
    Figure imgb0720
    Figure imgb0721
    Figure imgb0722
    when R3 is
    Figure imgb0723
    and R4 is halogen, CN, C1-C3 alkyl, C1-C3 haloalkyl or heteroaryl; or, when R3 is
    Figure imgb0724
    Figure imgb0725
    Figure imgb0726
    or
    Figure imgb0727
    , R5 is:
    Figure imgb0728
    Figure imgb0729
    Figure imgb0730
    Figure imgb0731
    Figure imgb0732
    or
    Figure imgb0733
    wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rh is
    Figure imgb0734
    Figure imgb0735
    Figure imgb0736
    or
    Figure imgb0737
    Ri is H, halogen, methyl or cyano.
  2. The compound according to claim 1, wherein in the general formula (1), R1 is H, F, Cl, Me, Et, isopropyl, vinyl, ethynyl or cyclopropyl.
  3. The compound according to claim 1 or 2, wherein in the general formula (1), R2 is CH3O-, CH3CH2O-, CF3CH2O-, CHF2CH2O-,
    Figure imgb0738
    or
    Figure imgb0739
  4. The compound according to any one of claims 1-3 wherein in the general formula (1), R3 is
    Figure imgb0740
    Figure imgb0741
    Figure imgb0742
    Figure imgb0743
    Figure imgb0744
  5. The compound according to any one of claims 1-4, wherein in the general formula (1), R4 is H, F, CN, Me, CF3,
    Figure imgb0745
  6. The compound according to any one of claims 1-5, wherein in the general formula (1), when R3 is
    Figure imgb0746
    , and R4 is H, R5 is:
    Figure imgb0747
    Figure imgb0748
    Figure imgb0749
    Figure imgb0750
    Figure imgb0751
    Figure imgb0752
    Figure imgb0753
    Figure imgb0754
    Figure imgb0755
    Figure imgb0756
    Figure imgb0757
    Figure imgb0758
    Figure imgb0759
    Figure imgb0760
    Figure imgb0761
    Figure imgb0762
    Figure imgb0763
    Figure imgb0764
    Figure imgb0765
    Figure imgb0766
    Figure imgb0767
    Figure imgb0768
    Figure imgb0769
    Figure imgb0770
  7. The compound according to any one of claims 1-5, wherein in the general formula (1), when R3 is
    Figure imgb0771
    , and R4 is F, CN, Me, CF3,
    Figure imgb0772
    ; or, when R3 is
    Figure imgb0773
    Figure imgb0774
    Figure imgb0775
    Figure imgb0776
    R5 is:
    Figure imgb0777
    Figure imgb0778
    Figure imgb0779
    Figure imgb0780
    Figure imgb0781
    Figure imgb0782
    Figure imgb0783
    Figure imgb0784
    Figure imgb0785
    Figure imgb0786
    Figure imgb0787
    Figure imgb0788
    Figure imgb0789
    Figure imgb0790
    Figure imgb0791
    Figure imgb0792
    Figure imgb0793
    Figure imgb0794
    Figure imgb0795
    Figure imgb0796
    Figure imgb0797
    Figure imgb0798
    Figure imgb0799
    Figure imgb0800
    Figure imgb0801
    Figure imgb0802
    Figure imgb0803
    Figure imgb0804
    Figure imgb0805
    Figure imgb0806
  8. The compound according to any one of claims 1-5, wherein in the general formula (1), R5 is:
    Figure imgb0807
    Figure imgb0808
    Figure imgb0809
    Figure imgb0810
    Figure imgb0811
    Figure imgb0812
    Figure imgb0813
    Figure imgb0814
    Figure imgb0815
    Figure imgb0816
    Figure imgb0817
    Figure imgb0818
    Figure imgb0819
    Figure imgb0820
    Figure imgb0821
  9. The compound, the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof according to any one of claims 1-8, wherein the compound has one of the following structures:
    Figure imgb0822
    Figure imgb0823
    Figure imgb0824
    Figure imgb0825
    Figure imgb0826
    Figure imgb0827
    Figure imgb0828
    Figure imgb0829
    Figure imgb0830
    Figure imgb0831
    Figure imgb0832
    Figure imgb0833
    Figure imgb0834
    Figure imgb0835
    Figure imgb0836
    Figure imgb0837
    Figure imgb0838
    Figure imgb0839
    Figure imgb0840
    Figure imgb0841
    Figure imgb0842
    Figure imgb0843
    Figure imgb0844
    Figure imgb0845
    Figure imgb0846
    Figure imgb0847
    Figure imgb0848
    Figure imgb0849
    Figure imgb0850
    Figure imgb0851
    Figure imgb0852
    Figure imgb0853
    Figure imgb0854
    Figure imgb0855
    Figure imgb0856
    Figure imgb0857
    Figure imgb0858
    Figure imgb0859
    Figure imgb0860
    Figure imgb0861
    Figure imgb0862
    Figure imgb0863
    Figure imgb0864
    Figure imgb0865
    Figure imgb0866
    Figure imgb0867
    Figure imgb0868
    Figure imgb0869
    Figure imgb0870
    Figure imgb0871
    Figure imgb0872
    Figure imgb0873
    Figure imgb0874
    Figure imgb0875
    Figure imgb0876
    Figure imgb0877
    Figure imgb0878
    Figure imgb0879
    Figure imgb0880
    Figure imgb0881
    Figure imgb0882
    Figure imgb0883
    Figure imgb0884
    Figure imgb0885
    Figure imgb0886
    Figure imgb0887
    Figure imgb0888
    Figure imgb0889
    Figure imgb0890
    Figure imgb0891
    Figure imgb0892
    Figure imgb0893
    Figure imgb0894
    Figure imgb0895
    Figure imgb0896
  10. A compound with a structure as shown in general formula (2), isomers thereof, crystalline forms thereof pharmaceutically acceptable salts thereof hydrates thereof or solvates thereof:
    Figure imgb0897
    wherein in general formula (2):
    R1a is
    Figure imgb0898
    R2a is CH3O-, CH3CH2O-, CF3CH2O- or CHF2CH2O-;
    R3ais
    Figure imgb0899
    Figure imgb0900
    wherein Rc is H or F, Rd is H, F, Cl or Me, Re is H, F, Cl or Me, and Rf is F, NH2, Me or cyclopropyl;
    R4a is H or F; and
    R5a is: H,
    Figure imgb0901
    Figure imgb0902
    Figure imgb0903
    or
    Figure imgb0904
    wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; v is an integer of 1, 2 or 3; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rj is independently halogen, CN, SO2Me, C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl or
    Figure imgb0905
    Rk is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy, C3-C6 cycloalkyl or
    Figure imgb0906
    Rn is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy or C3-C6 cycloalkyl, two Rn groups, together with one carbon atom, form a spiro ring, or two Rn groups, together with different carbon atoms, form a bridged ring; R1 and Rm are independently C1-C3 alkyl, C1-C3 haloalkyl, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, or R1 and Rm, together with a N atom, form a 3-8 membered heterocycloalkyl group, wherein the 3-8 membered heterocycloalkyl group may be substituted with 1-3 groups selected from OH, halogen, cyano, C1-C3 alkyl, C3-C6 cycloalkyl, heterocycloalkyl, (C1-C3)alkoxy or (halogenated C1-C3)alkoxy.
  11. The compound according to claim 10, wherein in the general formula (2), R3a is
    Figure imgb0907
    Figure imgb0908
    Figure imgb0909
  12. The compound according to claim 10 or 11, wherein in the general formula (2), R5a is: H,
    Figure imgb0910
    Figure imgb0911
    Figure imgb0912
    Figure imgb0913
    Figure imgb0914
    Figure imgb0915
    Figure imgb0916
    Figure imgb0917
    Figure imgb0918
    Figure imgb0919
    Figure imgb0920
    Figure imgb0921
    Figure imgb0922
    Figure imgb0923
    Figure imgb0924
    Figure imgb0925
    Figure imgb0926
    Figure imgb0927
    Figure imgb0928
    Figure imgb0929
    Figure imgb0930
    Figure imgb0931
    Figure imgb0932
    Figure imgb0933
    Figure imgb0934
    Figure imgb0935
    Figure imgb0936
    Figure imgb0937
    Figure imgb0938
    Figure imgb0939
    or
    Figure imgb0940
  13. The compound, the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof according to any one of claims 10-12, wherein the compound has one of the following structures:
    Figure imgb0941
    Figure imgb0942
    Figure imgb0943
    Figure imgb0944
    Figure imgb0945
    Figure imgb0946
    Figure imgb0947
    Figure imgb0948
    Figure imgb0949
    Figure imgb0950
    Figure imgb0951
    Figure imgb0952
    Figure imgb0953
    Figure imgb0954
    Figure imgb0955
    Figure imgb0956
    Figure imgb0957
    Figure imgb0958
    Figure imgb0959
    Figure imgb0960
    Figure imgb0961
    Figure imgb0962
    Figure imgb0963
    Figure imgb0964
    Figure imgb0965
    Figure imgb0966
    Figure imgb0967
    Figure imgb0968
    Figure imgb0969
    Figure imgb0970
    Figure imgb0971
    Figure imgb0972
    Figure imgb0973
    Figure imgb0974
    Figure imgb0975
    Figure imgb0976
    Figure imgb0977
    Figure imgb0978
    Figure imgb0979
  14. A compound with a structure as shown in general formula (3), isomers thereof, crystalline forms thereof pharmaceutically acceptable salts thereof hydrates thereof or solvates thereof:
    Figure imgb0980
    wherein, R5b is:
    Figure imgb0981
    Figure imgb0982
    Figure imgb0983
    Figure imgb0984
    Figure imgb0985
    , wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rh is
    Figure imgb0986
    Figure imgb0987
    Figure imgb0988
    Ri is H, halogen, methyl or cyano; or
    R5b is: H,
    Figure imgb0989
    Figure imgb0990
    Figure imgb0991
    Figure imgb0992
    , wherein n1, n2, n3, m1, m2 and m3 are independently integers of 1 or 2; v is an integer of 1, 2 or 3; Rg is C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, heterocycloalkyl, heterocycloalkyl-(C1-C3)alkyl-, C1-C3 haloalkyl or cyano-substituted C1-C3 alkyl; Rj is independently halogen, CN, SO2Me, C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl or
    Figure imgb0993
    Rk is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy, C3-C6 cycloalkyl or
    Figure imgb0994
    Rn is independently halogen, CN, OH, C1-C3 alkyl, C1-C3 alkoxy or C3-C6 cycloalkyl, two Rn groups, together with one carbon atom, form a spiro ring, or two Rn groups, together with different carbon atoms, form a bridged ring; R1 and Rm are independently C1-C3 alkyl, C1-C3 haloalkyl, hydroxy-substituted C1-C3 alkyl, cyano-substituted C1-C3 alkyl, C3-C6 cycloalkyl, (C1-C3)alkoxy-(C2-C3)alkyl-, (halogenated C1-C3)alkoxy-(C2-C3)alkyl-, (C3-C6)cycloalkyl-(C1-C3)alkyl-, or R1 and Rm, together with a N atom, form a 3-8 membered heterocycloalkyl group, wherein the 3-8 membered heterocycloalkyl group may be substituted with 1-3 groups selected from OH, halogen, cyano, C1-C3 alkyl, C3-C6 cycloalkyl, heterocycloalkyl, (C1-C3)alkoxy or (halogenated C1-C3)alkoxy.
  15. The compound according to claim 14, wherein
    Figure imgb0995
    Figure imgb0996
    Figure imgb0997
    Figure imgb0998
    Figure imgb0999
    Figure imgb1000
    Figure imgb1001
    Figure imgb1002
    Figure imgb1003
    Figure imgb1004
    Figure imgb1005
    Figure imgb1006
    Figure imgb1007
    Figure imgb1008
    Figure imgb1009
    Figure imgb1010
    Figure imgb1011
    Figure imgb1012
    Figure imgb1013
    Figure imgb1014
    Figure imgb1015
    Figure imgb1016
    Figure imgb1017
    Figure imgb1018
    Figure imgb1019
    Figure imgb1020
    Figure imgb1021
    Figure imgb1022
    Figure imgb1023
    Figure imgb1024
    Figure imgb1025
    Figure imgb1026
    Figure imgb1027
    Figure imgb1028
    Figure imgb1029
    Figure imgb1030
    Figure imgb1031
    Figure imgb1032
    Figure imgb1033
    Figure imgb1034
    Figure imgb1035
    Figure imgb1036
    Figure imgb1037
    Figure imgb1038
    Figure imgb1039
    Figure imgb1040
    Figure imgb1041
    Figure imgb1042
    Figure imgb1043
    Figure imgb1044
    Figure imgb1045
    Figure imgb1046
    Figure imgb1047
    Figure imgb1048
    Figure imgb1049
    Figure imgb1050
    Figure imgb1051
    Figure imgb1052
    Figure imgb1053
    Figure imgb1054
    Figure imgb1055
    Figure imgb1056
    Figure imgb1057
    Figure imgb1058
    Figure imgb1059
    Figure imgb1060
    Figure imgb1061
    Figure imgb1062
    Figure imgb1063
    Figure imgb1064
    Figure imgb1065
    Figure imgb1066
    Figure imgb1067
    Figure imgb1068
    or
    Figure imgb1069
  16. A pharmaceutical composition comprising a pharmaceutically acceptable excipient or carrier, and the compounds of general formulas (1) through (3), the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof according to any one of claims 1-15 as active ingredients.
  17. Use of the compounds of general formulas (1) through (3), the isomers thereof, the crystalline forms thereof, the pharmaceutically acceptable salts thereof, the hydrates thereof or the solvates thereof according to any one of claims 1-15 in preparing a medicament for treating RAS-associated diseases.
EP20905322.2A 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound Pending EP4083042A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201911386239.6A CN113045570A (en) 2019-12-27 2019-12-27 Spiro-containing quinazoline compounds
CN202010486384.8A CN113754659A (en) 2020-06-01 2020-06-01 Spiro-containing quinazoline compounds
PCT/CN2020/139530 WO2021129820A1 (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound

Publications (1)

Publication Number Publication Date
EP4083042A1 true EP4083042A1 (en) 2022-11-02

Family

ID=76573713

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20905322.2A Pending EP4083042A1 (en) 2019-12-27 2020-12-25 Spiro ring-containing quinazoline compound

Country Status (10)

Country Link
US (1) US20230002382A1 (en)
EP (1) EP4083042A1 (en)
JP (1) JP2023508482A (en)
KR (1) KR20220122662A (en)
CN (1) CN114929704A (en)
AU (1) AU2020414943A1 (en)
BR (1) BR112022010267A2 (en)
CA (1) CA3162106A1 (en)
MX (1) MX2022006609A (en)
WO (1) WO2021129820A1 (en)

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11932633B2 (en) 2018-05-07 2024-03-19 Mirati Therapeutics, Inc. KRas G12C inhibitors
JP2022517222A (en) 2019-01-10 2022-03-07 ミラティ セラピューティクス, インコーポレイテッド KRAS G12C inhibitor
JP2022546043A (en) 2019-08-29 2022-11-02 ミラティ セラピューティクス, インコーポレイテッド KRAS G12D inhibitor
CA3152025A1 (en) 2019-09-24 2021-04-01 David BRIERE Combination therapies
EP4076418A4 (en) 2019-12-20 2024-01-24 Mirati Therapeutics Inc Sos1 inhibitors
IL301062A (en) 2020-09-03 2023-05-01 Revolution Medicines Inc Use of sos1 inhibitors to treat malignancies with shp2 mutations
PE20231207A1 (en) 2020-09-15 2023-08-17 Revolution Medicines Inc INDOLIC DERIVATIVES AS RAS INHIBITORS IN CANCER TREATMENT
US20240109893A1 (en) * 2020-12-22 2024-04-04 Shanghai Kechow Pharma, Inc. Preparation and application method of heterocyclic compounds as kras inhibitor
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
IL310291A (en) * 2021-07-23 2024-03-01 Theras Inc Compositions and methods for inhibition of ras
WO2023008462A1 (en) 2021-07-27 2023-02-02 東レ株式会社 Medicament for treatment and/or prevention of cancer
AR127308A1 (en) 2021-10-08 2024-01-10 Revolution Medicines Inc RAS INHIBITORS
WO2023114954A1 (en) 2021-12-17 2023-06-22 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors
WO2024081674A1 (en) 2022-10-11 2024-04-18 Aadi Bioscience, Inc. Combination therapies for the treatment of cancer
WO2024102421A2 (en) 2022-11-09 2024-05-16 Revolution Medicines, Inc. Compounds, complexes, and methods for their preparation and of their use

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013159698A1 (en) * 2012-04-26 2013-10-31 深圳信立泰药业股份有限公司 Fused ring quinazoline hydroximic acid compound and use thereof as anti-tumour drug
CN103772411B (en) * 2013-04-28 2016-08-03 复旦大学 Sulfur-bearing 6-furyl quinazoline-4-amines and its production and use
US10414757B2 (en) * 2015-11-16 2019-09-17 Araxes Pharma Llc 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
CN110088098B (en) * 2016-01-26 2022-04-15 中国医学科学院药物研究所 Quinazolinone PARP-1 inhibitor and preparation method, pharmaceutical composition and application thereof
JOP20190186A1 (en) * 2017-02-02 2019-08-01 Astellas Pharma Inc Quinazoline compound
JP2021176819A (en) * 2018-07-31 2021-11-11 アステラス製薬株式会社 Pharmaceutical composition comprising quinazoline compound as active ingredient
JP2021176820A (en) * 2018-07-31 2021-11-11 アステラス製薬株式会社 Pharmaceutical composition comprising quinazoline compound as active ingredient
KR20210097715A (en) * 2018-11-29 2021-08-09 아락세스 파마 엘엘씨 Compounds for the treatment of cancer and methods of use thereof
CN111499634B (en) * 2019-01-31 2023-05-12 贝达药业股份有限公司 Quinazoline compound and application thereof in medicine
WO2020177629A1 (en) * 2019-03-01 2020-09-10 劲方医药科技(上海)有限公司 Spiro-substituted pyrimidine-fused cyclic compound, preparation method therefor and medical use thereof
CN112110918B (en) * 2019-06-21 2023-08-22 劲方医药科技(上海)有限公司 Spiro substituted pyrimido cyclic compounds, process for their preparation and their use in medicine

Also Published As

Publication number Publication date
CN114929704A (en) 2022-08-19
US20230002382A1 (en) 2023-01-05
WO2021129820A1 (en) 2021-07-01
JP2023508482A (en) 2023-03-02
MX2022006609A (en) 2022-07-05
BR112022010267A2 (en) 2022-08-16
CA3162106A1 (en) 2021-07-01
AU2020414943A1 (en) 2022-08-04
KR20220122662A (en) 2022-09-02

Similar Documents

Publication Publication Date Title
EP4083042A1 (en) Spiro ring-containing quinazoline compound
CN115335379B (en) Spirocyclic quinazoline compounds
CN113767103B (en) Novel spirocyclic K-Ras G12C inhibitors
WO2021129824A1 (en) New-type k-ras g12c inhibitor
EP2947086B1 (en) Novel fused pyrimidine compound or salt thereof
WO2020259432A1 (en) Kras-g12c inhibitor
CA3213029A1 (en) Parp inhibitor containing piperazine structure, preparation method therefor and pharmaceutical use thereof
CN116390728B (en) Quinazoline derivative, preparation method and application thereof
CN113045570A (en) Spiro-containing quinazoline compounds
CN114736214B (en) Sesquiterpene derivative, pharmaceutical composition thereof, and preparation method and application thereof
EP4265620A1 (en) Novel camptothecin derivative, composition containing same, and use thereof
CN111646986B (en) Nicotinic acid derivative and preparation method and application thereof
CN113754659A (en) Spiro-containing quinazoline compounds
KR102606167B1 (en) Fluorine-containing substituted benzothiophene compounds, pharmaceutical compositions and applications thereof
CN113880804A (en) Novel benzimidazole compounds
CN107163047B (en) Sophoridine amine derivative and preparation method and application thereof
CN116783194A (en) Spirocyclic quinazoline derivatives
EP4159731A1 (en) New pyrazine compound
CN109942566B (en) Isonicotinic acid derivative and preparation method and application thereof
KR101071359B1 (en) 2-methyl-2-alkyl-6-amido-2h-benzopyran derivatives having anti-cancer activity
EP4332104A1 (en) Fused ring compound as wee-1 inhibitor, and preparation method therefor and use thereof
TW202306955A (en) Naphthyridine derivative as ATR inhibitor and method for preparing same
CN116693470A (en) Histamine H3 receptor inhibitor and medical application thereof

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20220727

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 35/00 20060101ALI20240219BHEP

Ipc: A61K 31/517 20060101ALI20240219BHEP

Ipc: C07D 471/10 20060101AFI20240219BHEP